EP1578748B1 - Tetrahydro-4h-pyrido[1,2-a]pyrimidines and related compounds useful as hiv integrase inhibitors - Google Patents

Tetrahydro-4h-pyrido[1,2-a]pyrimidines and related compounds useful as hiv integrase inhibitors Download PDF

Info

Publication number
EP1578748B1
EP1578748B1 EP03768014A EP03768014A EP1578748B1 EP 1578748 B1 EP1578748 B1 EP 1578748B1 EP 03768014 A EP03768014 A EP 03768014A EP 03768014 A EP03768014 A EP 03768014A EP 1578748 B1 EP1578748 B1 EP 1578748B1
Authority
EP
European Patent Office
Prior art keywords
oxo
hydroxy
amino
fluorobenzyl
tetrahydro
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
EP03768014A
Other languages
German (de)
French (fr)
Other versions
EP1578748A1 (en
Inventor
Benedetta IRBM CRESCENZI
Olaf IRBM KINZEL
Ester IRBM MURAGLIA
Federica IRBM ORVIETO
Giovanna IRBM PESCATORE
Michael IRBM ROWLEY
Vincenzo IRBM SUMMA
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Istituto di Ricerche di Biologia Molecolare P Angeletti SpA
Original Assignee
Istituto di Ricerche di Biologia Molecolare P Angeletti SpA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Istituto di Ricerche di Biologia Molecolare P Angeletti SpA filed Critical Istituto di Ricerche di Biologia Molecolare P Angeletti SpA
Publication of EP1578748A1 publication Critical patent/EP1578748A1/en
Application granted granted Critical
Publication of EP1578748B1 publication Critical patent/EP1578748B1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present invention is directed to tetrahydro-4H-pyrido[1,2-a]pyrimidines, related compounds, and pharmaceutically acceptable salts thereof, their synthesis, and their use as inhibitors of the HIV integrase enzyme.
  • the compounds and pharmaceutically acceptable salts thereof of the present invention are useful for preventing or treating infection by HIV and for treating or delaying the onset of AIDS.
  • a retrovirus designates human immunodeficiency virus (HIV) is the etiological agent of the complex disease that includes progressive destruction of the immune system (acquired immune deficiency syndrome; AIDS) and degeneration of the central and peripheral nervous system.
  • This virus was previously known as LAV, HTLV-III, or ARV.
  • a common feature of retrovirus replication is the insertion by virally-encoded integrase of proviral DNA into the host cell genome, a required step in HIV replication in human T-lymphoid and monocytoid cells.
  • Integration is believed to be mediated by integrase in three steps: assembly of a stable nucleoprotein complex with viral DNA sequences; cleavage of two nucleotides from the 3' termini of the linear proviral DNA; covalent joining of the recessed 3' OH termini of the proviral DNA at a staggered cut made at the host target site.
  • the fourth step in the process, repair synthesis of the resultant gap may be accomplished by cellular enzymes.
  • Nucleotide sequencing of HIV shows the presence of a pol gene in one open reading frame [ Ratner, L. et al., Nature, 313, 277(1985 )].
  • Amino acid sequence homology provides evidence that the pol sequence encodes reverse transcriptase, integrase and an HIV protease [ Toh, H. et al., EMBO J. 4, 1267 (1985 ); Power, M.D. et al., Science, 231,1567 (1986 ); Pearl, L.H et al., Nature, 329, 351 (1987 )]. All three enzymes have been shown to be essential for the replication of HIV.
  • antiviral compounds which act as inhibitors of HIV replication are effective agents in the treatment of AIDS and similar diseases, including reverse transcriptase inhibitors such as azidothymidine (AZT) and efavirenz and protease inhbitors such as indinavir and nelfinavir.
  • the compounds of this invention are inhibitors of HIV integrase and inhibitors of HIV replication.
  • the inhibition of integrase in vitro and HIV replication in cells is a direct result of inhibiting the strand transfer reaction catalyzed by the recombinant integrase in vitro in HIV infected cells.
  • the particular advantage of the present invention is highly specific inhibition of HIV integrase and HIV replication.
  • the present invention is directed to novel pyridopyrimidine derivatives and related compounds. These compounds are useful in the inhibition of HIV integrase, the prevention of infection by HIV, the treatment of infection by HIV and in the prevention, treatment, and delay in the onset of AIDS and/or ARC, either as compounds or their pharmaceutically acceptable salts or hydrates (when appropriate), or as pharmaceutical composition ingredients, whether or not in combination with other HIV/AIDS antivirals, anti-infectives, immunomodulators, antibiotics or vaccines.
  • the present invention also includes pharmaceutical compositions containing a compound of the present invention and methods of preparing such pharmaceutical compositions.
  • the present invention further includes the use of the compounds for treating AIDS, delaying the onset of AIDS, preventing AIDS, preventing infection by HIV, and treating infection by HIV.
  • R 1 is NR 2 R 5 ;
  • R 2 is CH 3 ;
  • R 5 is 1) C(O)CH 2 SO 2 CH 3 , 2) C(O)C(O)N(CH 3 ) 2 , 3) SO 2 N(CH 3 ) 2 , or 4) SO 2 R 20 , wherein R 20 is: or alternatively R 2 and R 5 together with the nitrogen atom to which they are attached form or R3 is hydrogen;
  • R 4 is p-fluorobenzyl, 4-fluoro-3-methylbenzyl, 3-chlorobenzyl, or 3-chloro-4-methylbenzyl;
  • R 12 and R 14 are both H, except that when R 5 is C(O)C(O)N(CH 3 ) 2 and R 4 is p-fluorobenzyl and n is 1, then R 12 and R 14 are either both H or both CH 3 ; and n is an integer equal to 1 or 2.
  • R 1 is NR 2 R 5 ;
  • R 2 is CH 3 ;
  • R 5 is 1) C(O)C(O)N(CH 3 ) 2 or 2) SO 2 R 20 , wherein R 20 is R 3 is hydrogen;
  • R 4 is p-fluorobenzyl or 4-fluoro-3-methylbenzyl;
  • R 12 and R 14 are both H, except that when R 5 is C(O)C(O)N(CH 3 ) 2 and R 4 is p-fluorobenzyl and n is 1, then R 12 and R 14 are either both H or both CH 3 ; and n is an integer equal to 1 or 2.
  • An embodiment of the present invention is a compound, or a pharmaceutically acceptable salt thereof, selected from the group consisting of the compounds set forth in Table 1 below.
  • the present invention also includes a compound of the present invention (i) for use in, (ii) for use as a medicament for, or (iii) for use in the preparation of a medicament for (a) inhibiting HTV integrase, (b) preventing or treating infection by HIV, or (c) preventing, treating or delaying the onset of AIDS.
  • the compounds of the present invention can optionally be employed in combination with one or more HIV/AIDS treatment agents selected from HIV/AIDS antiviral agents, anti-infective agents, and immunomodulators.
  • Additional embodiments of the invention include the pharmaceutical compositions, combinations and methods set forth in (a)-(i) above and the uses set forth in the preceding paragraph, wherein the compound of the present invention employed therein is a compound of one of the embodiments, aspects, classes, sub-classes, or features of the compounds described above. In all of these embodiments, the compound may optionally be used in the form of a pharmaceutically acceptable salt.
  • the compounds of the present invention may have asymmetric centers and may occur, except when specifically noted, as mixtures of stereoisomers or as individual diastereomers, or enantiomers, with all isomeric forms being included in the present invention.
  • N-substituted hydroxypyrimidinone compounds of the present invention may also occur as tautomers thereof, such as the following tautomer of a compound of Formula II: It is understood that the present invention includes all tautomers of the hydroxypyrimidinone compounds of Formula II, both singly and in mixtures.
  • the compounds of the present inventions are useful in the inhibition of HIV integrase, the prevention or treatment of infection by human immunodeficiency virus (HIV) and the prevention, treatment or the delay in the onset of consequent pathological conditions such as AIDS.
  • HIV human immunodeficiency virus
  • Preventing AIDS, treating AIDS, delaying the onset of AIDS, or preventing or treating infection by HIV is defined as including, but not limited to, treatment of a wide range of states of HIV infection: AIDS, ARC (AIDS related complex), both symptomatic and asymptomatic, and actual or potential exposure to HIV.
  • the compounds of this invention are useful in treating infection by HIV after suspected past exposure to HIV by such means as blood transfusion, exchange of body fluids, bites, accidental needle stick, or exposure to patient blood during surgery.
  • the compounds of this invention are useful in the preparation and execution of screening assays for antiviral compounds.
  • the compounds of this invention are useful for isolating enzyme mutants, which are excellent screening tools for more powerful antiviral compounds.
  • the compounds of this invention are useful in establishing or determining the binding site of other antivirals to HIV integrase, e.g., by competitive inhibition.
  • the compounds of this invention are commercial products to be sold for these purposes.
  • Certain compounds representative of the present invention have also been tested in an assay for inhibition of acute HIV infection of T-lymphoid cells, conducted in accordance with Vacca, J.P. et al., Proc. Natl. Acad. Sci. USA 1994, 91: 4096 . These compounds-including the compounds set forth below in Table 1- demonstrated IC 95 's of about 20 micromolar or less.
  • the compounds of the present invention can also act as inhibitors of HIV ribonuclease H (RNase H).
  • RNase H The human immunodeficiency virus type 1 (HIV-1) reverse transcriptase (RT) catalyzes the conversion of genomic RNA into double-stranded proviral DNA after cell entry, utilizing the RNA- and DNA-dependent polymerase and RNase H activities of the enzyme.
  • HIV-1 RT is an asymmetric dimer consisting of p66 and p51 polypeptides.
  • the catalytic activities of RT are conducted at discrete sites in the p66 subunit; i.e., the N terminus of p66 catalyzes the RNA- and DNA-dependent DNA polymerase activity, an the p15 domain at the C terminus catalyzes RNase H activity.
  • RNase H is required to cleave the RNA strand of the RNA:DNA heteroduplex intermediates in reverse transcription.
  • the compounds of the present invention can selectively bind to and inhibit the RNase H domain of HIV-1 RT.
  • the RNase H inhibition activity of the compounds can be measured using suitable assays known in the art, such as the assay described in Shaw-Reid et al., J. Biol. Chem.
  • the present invention includes a method of inhibiting HIV RNase H in a subject in need of such inhibition which comprises administering to the subject an effective amount of a compound of the invention.
  • the present invention further includes a compound of the present invention (i) for use in, (ii) for use as a medicament for, or (iii) for use in the preparation of a medicament for inhibiting HIV RNase H.
  • the compounds of the present invention may be administered in the form of pharmaceutically acceptable salts.
  • pharmaceutically acceptable salt refers to a salt which possesses the effectiveness of the parent compound and which is not biologically or otherwise undesirable (e.g., is neither toxic nor otherwise deleterious to the recipient thereof).
  • Suitable salts include acid addition salts which may, for example, be formed by mixing a solution of the compound of the present invention with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulfuric acid, acetic acid, trifluoroacetic acid, or benzoic acid
  • suitable pharmaceutically acceptable salts thereof can include alkali metal salts (e.g., sodium or potassium salts), alkaline earth metal salts (e.g., calcium or magnesium salts); and salts formed with suitable organic ligands such as quaternary ammonium salts.
  • alkali metal salts e.g., sodium or potassium salts
  • alkaline earth metal salts e.g., calcium or magnesium salts
  • suitable organic ligands such as quaternary ammonium salts.
  • pharmaceutically acceptable esters can be employed to modify the solubility or hydrolysis characteristics of the compound.
  • the compounds of the present invention may be administered orally, parenterally (including subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques), by inhalation spray, or rectally, in the form of a unit dosage of a pharmaceutical composition containing a therapeutically effective amount bf the compound and conventional non-toxic pharmaceutically-acceptable carriers, adjuvants and vehicles.
  • administration and variants thereof (e.g., “administering” a compound) in reference to a compound of the invention mean providing the compound or a prodrug of the compound to the individual in need of treatment.
  • a compound of the invention or a prodrug thereof is provided in combination with one or more other active agents (e.g., antiviral agents useful for treating HIV infection or AIDS)
  • “administration” and its variants are each understood to include concurrent and sequential provision of the compound or prodrug and other agents.
  • composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combining the specified ingredients in the specified amounts.
  • pharmaceutically acceptable is meant that the ingredients of the pharmaceutical composition must be compatible with each other and not deleterious to the recipient thereof.
  • subject refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
  • terapéuticaally effective amount means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation or prophylaxis of the symptoms of the disease or condition being treated or prevented.
  • the term also includes herein the amount of active compound sufficient to inhibit HIV integrase and/or RNase H and thereby elicit the response being sought.
  • references to the amount of active ingredient are to the free acid or free base form of the compound.
  • compositions may be in the form of orally-administrable suspension or tablets or capsules, nasal sprays, sterile injectible preparations, for example, as sterile injectible aqueous or oleagenous suspensions or suppositories.
  • sterile injectible preparations for example, as sterile injectible aqueous or oleagenous suspensions or suppositories.
  • These compositions can be prepared by methods and contain excipients which are well known in the art. Suitable methods and ingredients are described in Remington's Pharmaceutical Sciences, 18th edition, edited by A. R. Gennaro, Mack Publishing Co.,1990 , which is herein incorporated by reference in its entirety.
  • the compounds of this invention can be administered orally in a dosage range of 0.001 to 1000 mg/kg of mammal (e.g., human) body weight per day in a single dose or in divided doses.
  • mammal e.g., human
  • One referred dosage range is 0.01 to 500 mg/kg body weight per day orally in a single dose or in divided doses.
  • Another preferred dosage range is 0.1 to 100 mg/kg body weight per day orally in single or divided doses.
  • the compositions can be provided in the form of tablets or capsules containing 1.0 to 500 milligrams of the active ingredient, particularly 1, 5,10,15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
  • the present invention is also directed to use of the HIV integrase inhibitor compounds of the present invention with one or more agents useful in the treatment of HIV infection or AIDS.
  • the compounds of this invention may be effectively administered, whether at periods of pre-exposure and/or post-exposure, in combination with effective amounts of one or more HIV/AIDS antivirals, imunomodulators, antiinfectives, or vaccines useful for treating HIV infection or AIDS, such as those disclosed in Table 1 of WO 01/38332 or in the Table in WO 02/30930 , both documents being herein incorporated by reference in their entireties.
  • AIDS acquired immunodeficiency syndrome
  • ARC AIDS related complex
  • DBU 1,8-diazabicyclo[5.4.0]undec-7-ene
  • DMAD dimethylacetylenedicarboxylate
  • DMF N,N-dimethylformamide
  • DMSO dimethylsulfoxide
  • EtOAc ethyl acetate
  • FIA-MS flow injection analysis mass spectrometry
  • h hour(s)
  • HIV human immunodeficiency virus
  • HPLC high performance liquid chromatography
  • IPA isopropanol
  • LDA lithium diisopropylamide
  • Me methyl
  • MeOH methanol
  • NMP N-methyl pyrrolidinone
  • NMR nuclear magnetic resonance
  • Pd/C palladium on carbon catalyst
  • the compounds of the present invention can be readily prepared according to the following reaction schemes and examples, or modifications thereof, using readily available starting materials and reagents. In the reactions shown below, it is also possible to make use of variants which are themselves known to those of ordinary skill in this art, but are not mentioned in greater detail. Furthermore, other methods for preparing compounds of the invention (including compounds embraced by Formula A and Formula I but whose preparation is not literally described below) will be readily apparent to the person of ordinary skill in the art in light of the following reaction schemes and examples. Unless otherwise defined, the variables listed in Schemes 1, A, B, C, D and E have the following meanings:
  • P ⁇ is hydrogen or a protective group, e.g., an ester such as, but not limited to, benzoate and pivalate, or an ether such as, but not limited to, a benzyl ether, that is normally removed under the conditions employed to convert the methyl ester to the amide or is removed in a different step.
  • the protective group is typically used for synthetic and/or purification reasons.
  • the compounds of the present invention can be prepared by coupling the appropriate amines with suitable substituted alkyl 3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H pyrido[1,2-a] pyrimidine -2-carboxylate (or carboxylic acids or halides) or alkyl 3-hydroxy-4-oxo-4,6,7,8-tetrahydropyrrolo[1,2-a]pyrimidine-2-carboxylate (or carboxylic acids or halides) or alkyl 3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxylate (or carboxylic acids or halides) or alkyl 3-hydroxy-4-oxo-6,7,8,9,10,11-hexahydro-4 H -pyrimido[1,2-a]azepine-2-carboxylate (or carboxylic acids or halides), as represented by Scheme 1.
  • Scheme A depicts a general synthesis of carboxamides A-5.
  • the methyl ester A-4 can be reacted with an amine 1-2 in solvents like DMF, methanol, ethanol, toluene, NMP at the appropriate temperature (e.g., from 20 to 150°C) to give the final compound A-5.
  • Scheme A is exemplified in Reference Examples 1 and 9.
  • Scheme B shows a method for preparing compounds of the present invention that contain an amine, ether, thioether, sulfoxide or sulfone group at the 9-position of the 3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide core.
  • the bromo derivative B-1 can be obtained from methyl ester A-4 by first protecting the hydroxy group on A-4 with a suitable protective group (e.g., conversion to a benzoate or pivalate or to benzyloxy) and then contacting the protected A-4 with a brominating agent (e.g., NBS).
  • a suitable protective group e.g., conversion to a benzoate or pivalate or to benzyloxy
  • the bromo derivative B-1 can then be treated with a nucleophile ("Nu"; e.g. an amine, thiol or alcoholate) to afford with or without isolation the methyl ester intermediate B-2, that is reacted with the desired amine to give the final product B-3.
  • a nucleophile e.g. an amine, thiol or alcoholate
  • an oxidation step to obtain the sulfoxide or sulfone can be included in the scheme.
  • the nucleophile contains an ester, the ester can be converted to an amide by routine chemistry after the synthesis of B-3.
  • Scheme B is exemplified in Example 2.
  • Scheme C depicts a general synthesis of derivatives C-3 or C-4 containing an aliphatic ring substituent such as amide, sulfonamide, sulfonylurea, carbamate, or urea.
  • the bromo derivative B-1 can be treated with benzylamine C-1 and then hydrogenated or reacted directly with amine C-1a to give intermediate C-2 , which can then be treated with amine 1-2, with or without isolation, and then coupled with a carboxylic acid or reacted with a carbonyl chloride (or sulfonyl chloride or sulfamoyl chloride) or isocyanate to afford the final product C-3 .
  • Scheme C is exemplified in Examples 3,4,10 and 11. The last two steps can be reversed.
  • Scheme D shows the synthesis of homochiral compounds C-3, C3a,b , and C-4.
  • the bromo derivative B-1 is displaced with chiral amines D-1 to give a mixture of diastereoisomers, with subsequent or simultaneous removal of the protecting group.
  • the amino group at the 9-position is reductively alkylated with aldehydes or ketones D-2 to afford the intermediate D-3.
  • the mixture of diastereoisomers can be separated by crystallization or chromatography to give the single diastereoisomers D-3a,b .
  • R s6 can be removed by hydrogenation to afford the homochiral C-2a,b intermediate.
  • Step 1a Tert -butyl benzyloxy(4-cyanobutyl)carbamate (Bergeron, R. J., McManis, J. S., Tetrahedron 45 (16), 4939-4944 (1989).
  • Step 4a Methyl 2-(2-methoxy-2-oxoethyl)-56,7,8-tetrahydro-2 H -[1,2,4]oxadiazolo[2,3- a ]pyridine-2-carboxylate
  • Step 5a Methyl 3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4 H -pyrido[1,2- a ]pyrimidine-2-carboxylate
  • Step 2b Methyl 3-(benzyloxy)4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate
  • Step 3b Methyl 3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate
  • Step 6 N -(4-Fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4 H -pyrido[1,2- a ]pyrimidine-2-carboxamide .
  • Step 1 Methyl 3-(benzoyloxy)-4-oxo-6,7,8,9-tetrahydro-4 H -pyrido[1,2- a ]pyiimidine-2-carboxylate
  • Step 2 Methyl 3-(benzoyloxy)-9-bromo-4-oxo-6,7,8,9-tetrahydro-4 H -pyrido[1,2- a ]pyrimidine-2-carboxylate
  • Step 3 N-(4-Fluorobenzyl)-3-hydroxy-9-morpholin-4-yl-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamido.
  • Step 1 Methyl 9-[benzyl(methyl)amino]-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate hydrochloride
  • Step 2 Methyl 3-hydroxy-9-(methylamino)-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate
  • Step 3 N -(4-Fluombenzyl)-3-hydroxy-9-(methylamino)-4-oxo-6,7,8,9-tetrahydro-4 H- pyrido[1,2- a ]pyrimidine-2-carboxamide
  • Step 4 (+/-)-9-[[(Dimemylamino)sulfonyl](methyl)amino]- N -(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2- a ]pyrimidine-2-carboxamide
  • Step 1 (+/-)- N 1 -(2- ⁇ [(4-Fluorobenzyl)amino]carbonyl ⁇ -3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)- N 1 , N 2 , N 2 -trimethylethanediamide
  • Step 1 (+)3-Hydroxy-2-(methoxycarbonyl)- N -methyl-4-oxo- N -[(1 S )-1-phenylethyl]-6,7,8,9-tetrahydro-4 H -pyrido[1,2- a ]pyrimidin-9-ammonium trifluoroacetate
  • Step 2 (-)-3-Hydroxy-2-(methoxycarbonyl)- N -methyl-4-oxo-6,7,8,9-tetrahydro-4 H- pyrido[1,2- a ]pyrimidin-9-ammonium trifluoroacetate
  • Step 3 (+)- N 1 -(2- ⁇ [(4-Fluorobenzyl)amino]carbonyl ⁇ -3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4 H -pyrido[1,2- a ]pyrimidin-9-yl)- N 1 , N 2 , N 2 -trimethylethanediamide
  • Step 1 (-)-3-Hydroxy-2-(methoxycarbonyl)- N -methyl-4-oxo- N -[(1 R )-1-phenylethyl]-6,7,8,9-tetrahydro-4 H -pyrido[1,2- a ]pyrimidin-9-ammonium trifluoroacetate
  • Step 2 (+)-3-Hydroxy-2-(methoxycarbonyl)- N -methyl-4-oxo-6,7,8,9-tetrahydro-4 H- pyrido[1,2- a ]pyrimidin-9-ammonium trifluoroacetate
  • Step 3 (-)- N 1 -(2- ⁇ [(4-Fluorobenzyl)amino]carbonyl ⁇ -3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4 H -pyrido[1,2-a]pyrimidin-9-yl)- N 1 , N 2 , N 2 -trimethylethanediamide
  • Step 1 (-)-9-[[(Dimethylamino)sulfonyl](methyl)amino]- N -(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4 H -pyrido[1,2- a ]pyrimidine-2-carboxamide
  • the aqueous phase was extracted with ethylacetate and the combined organic phases were dried over sodium sulfate, filtered and concentrated to dryness under reduced pressure.
  • the product was purified by preparative RP-HPLC. using water (0.1 % TFA) and acetonitrile (0.1% TFA) as eluents (column: C18). The pooled product fractions were lyophilized to afford the title product as a fluffy white material (ee 90.7 %).
  • the compound was dissolved in ethanol and left standing at room temperature for three days. The supernatant was taken off and concentrated to dryness under reduced pressure. The residue was redissolved in ethanol and the solution was left standing at room temperature for one day.
  • Step 1 (+)-9-[[(Dimethylamino)sulfonyl](methyl)amino]- N -(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4 H -pyrido[1,2- a ]pyrimidine-2-carboxamide (16b)
  • Example 7 The title compound was synthesized using as starting material the amine prepared in Example 6, Step 2 and following the procedure describe in Example 7, Step 1.
  • the compound was obtained directly with the enantiomeric excess reported below and with spectroscopical properties identical to the compound synthesized in Example 3, Step 4 except for the optical rotation.
  • Step 1 tert -butyl benzyloxy(3-cyanopropyl)carbamate (Bergeron, R. J., McManis, J. S., Tetrahedron 45 (16), 4939-4944 (1989)).
  • Step 4 Methyl 3-hydroxy-4-oxo-4,6,7,8-tetrahydropyrrolo[1,2- a ]pyrimidino-2-carboxylate.
  • Step 5 Methyl 3-(benzoyloxy)-4-oxo-6,7,8,9-tetrahydropyrrolo[1,2- a ]pyrimidine-2-carboxylate (A) and 4-oxo-4,6,7,8-tetahydropyrrolo[1,2-a]pyrimidin-3-yl benzoate (B)
  • Step 6 N -(4-fluorobenzyl)-3-hydroxy-4-oxo-4,6,7,8-tetrahydrapyrrolo[1,2- a ]pyrimidine-2-carboxamide.
  • Step 2 (+/-)- N -(2- ⁇ [(4-Fluorobenzyl)amino]carbonyl ⁇ -3-hydroxy-4-oxo-4-6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)- N , N ', N '-trimethylethanediamide
  • the pooled product fractions were lyophilized to afford the title compound as a fluffy, white material.
  • the product is a mixture of rotamers by 1 H NMR.
  • Step.1 Dimethyl (2 E )-2-[(azepan-2-ylidencamino)oxy]but-2-enedioate and dimethyl (2 Z )-2-[(azepan-2-ylideneamino)oxy]but-2-enedioate
  • Step 2 Methyl 3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2- a ]azepine-2-carboxylate
  • Step 3 Methyl 3-(benzoyloxy)-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2- a ]azepine-2-carboxylate
  • Step 4 Methyl 3-(benzoyloxy)-10-bromo-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2- a ]azepine-2-carboxylate
  • Step 5 Methyl 3-hydroxy-4-oxo-10- ⁇ [(1 R )-1-phenylethyl]amino ⁇ -4,6,7,8,9,10-hexahydropyrimido[1,2- a ]azepine-2-carboxylate
  • Step 5A solid methyl 3-(benzoyloxy)-10-bromo-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2- a ]azepine-2-carboxylate (1.0 eq.) was added to a solution of (1 R )-1-phenylethylamine (45 eq.) dissolved in a 7:3 methanol/water mixture at -30°C. The reaction was carried out over night, then the temperature was raised to room temperature and the solvent concentrated to obtain a white solid that was filtered off and discarded. The title compound ( as a mixture 7:3 of diasteroisomers) was extracted in dichloromethane from the mother liquid for use in the next step without further purification. MS m / z : 358 (M+H) + .
  • Step 6 N -(4-fluorobenzyl)-3-hydroxy-4-oxo-10- ⁇ [(1 R )-1-phenylethyl]amino ⁇ -4,6,7,8,9,10-hexahydropyrimido[1,2- a ]azepine-2-carboxamide
  • Step 7 (+) N -(4-fluorobenzyl)-3-hydroxy-10- ⁇ methyl[(1 R )-1-phenylethyl]amino ⁇ -4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2- a ]azepine-2-carboxamide
  • N -(4-Fluorobenzyl)-3-hydroxy4-oxo-10-([(1 R )-1-phenylethyl]amino)-4,6,7,8,9,10-hexahydropyrimido[1,2- a ]azepine-2-carboxamide obtained in Step 6 was dissolved in methanol and the pH was adjusted to circa 5 with acetic acid before 37 % aqueous formaldehyde (6 eq.) and sodium cyanoborohydride (6.25 eq.) were added. The mixture was stirred at room temperature overnight.
  • Step 7A the product of Step 6A was reacted in the same manner as the product of Step 6 to obtain a single diastereomer without separation by HPLC.
  • Step 8 (-)2- ⁇ [(4-fluorobenzyl)amino]carbonyl ⁇ -3-hydroxy- N -methyl-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-ammonium trifluoroacetate
  • Step 9 (-) N -(- ⁇ [(4-Fluorobenzyl)amino]carbonyl ⁇ -3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2- a ]azepin-10-yl)- N,N',N' -trimethylethanediamide methyl chlorooxoacetate (2-6 eq.) and N-ethyldiisopropylamine (4 eq.) were added to a solution of the ammonium tritfluoroacetate compound of Step 8 in chloroform. The mixture was stirred at room temperature for 1 hour.
  • HPLC conditions Column: Zorbax, Rx C8 250 x 4.6 mm; Temperature: 30 ° C; Detection at 210 nm; Mobile Phase: 0.1% aq H 3 PO 4 (A)/eoCN (B); Gradient: 90:10 (A)/(B) to 10:90 over 15 min, 10:90 hold for 5 min. 10:90 to 90:10 (A)/(B) over 10 seconds; Flow Rate: 1 mL/min. Retention time: amidoxime 6 - 12.051 minutes, 12.315 minutes, ratio ca 3.6: 1.
  • the resulting solution was aged at the same temperature for 6-10 h (monitored by HPLC).
  • the by-product (MeSO 3 Me) which was generated in 1 equiv from the selectively hydrolysis of the trimesyl-pynmidine, was removed by azeotrope with DMF at 60-65 °C (monitored by 1 H NMR until ⁇ 10 mole%).
  • the concentration of bismesyl-pyrimidine 8 in DMP was about 0.3 M (total volume 300 mL).
  • 1 H NMR (CDCl 3 , 400 MHz) ⁇ : 11.00 (br s, 1 H), 4.78 (d, J 7.8 Hz, 1 H), 4.24-4.15 (m, 2 H), 3.95 (s, 3 H), 3.50 (s, 3 H), 2.99 (s, 3H), 2.81 (s, 3 H), 2.12-2.11 (m, 1 H), 1.90-1.76 (m, 2 H), 1.46 (s, 9 H), 1.43-1.35 (m, 2 H).
  • HPLC conditions Column: Zorbax, Rx C8 250 x 4.6 mm; Temperature: 30 ° C; Detection at 210 nm; Mobile Phase: 0.1% aq H 3 PO 4 (A)/MeCN (B); Gradient: 90: 10 (A)/(B) to 10:90 over 15 min, 10:90 hold for 5 min, 10:90 to 90:10 (A)/(B) over 10 seconds; Flow Rate: 1 mL/min. Retention time: trimesyl-pyrimidine - 14.140 minutes; bismesyl-pyrimidine 12.760 minutes.
  • HPLC conditions Column: Zorbax, Rx C8 250 x 4.6 mm; Temperature: 30 ° C; Detection at 210 nm; Mobile Phase: 0.1% aq H 3 PO 4 (A)/MeCN (B); Gradient: 90:10 (A)/(B) to 10:90 over 15 min, 10:90 hold for 5 min, 10:90 to 90:10 (A)/(B) over 10 seconds; Flow Rate: 1 mL/min. Retention time: the seven-membered ring-pyrimidine 10 - 15.467 minutes.
  • HPLC conditions Column: Zorbax, Rx C8 250 x 4.6 mm; Temperature: 30 ° C; Detection at 210 nm; Mobile Phase: 0.1% aq H 3 PO 4 (A)/MeCN (B); Gradient: 90:10 (A)/(B) to 10:90 over 15 min, 10:90 hold for 5 min, 10:90 to 90:10 (A)/(B) over 10 seconds; Flow Rate: 1 mL/min. Retention time: the seven-membered ring-pyrimidine hydrochloride salt 11 - 8.118 minutes.
  • Dimethylamine aqueous (40%, 158 ⁇ l) was added to the reaction mixture, and the mixture aged at 10-15 °C for 1 h, wherein the reaction was monitored by HLPC to assure complete conversion.
  • EtOAc (6 mL) and brine (2 mL) were added, respectively.
  • the organic layer was washed with 1 N HCl (2 mL), brine (2 x 2 mL).
  • the organic layer was concentrated to a total volume of 1 mL. Hexane (5 mL) was slowly added over 0.5 h.
  • the resulting slurry was aged at 0-5 °C for 1 h.
  • HPLC conditions Column: Zorbax, Rx C8 250 x 4.6 mm; Temperature: 30 ° C; Detection at 210 nm; Mobile Phase: 0.1% aq H 3 PO 4 (A)/MeCN (B); Gradient: 90:10 (A)/(B) to 10:90 over 15 min, 10:90 hold for 5 min, 10:90 to 90:10 (A)/(B) over 10 seconds; Flow Rate: 1 mL/min. Retention time: the title compound 14 - 12.191 minutes.
  • Table 1 below lists Compounds of the present invention which have been prepared.
  • the table provides the structure and name of each compound, the mass of its molecular ion plus 1 (M+) or molecular ion minus 1 (M-) as determined via FIA-MS, and the synthetic scheme employed to prepare the compound.

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Virology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • AIDS & HIV (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

Tetrahydro-4H-pyrido[1,2-a]pyrimidines and related compounds of Formula A: are described as inhibitors of HIV integrase and inhibitors of HIV replication, wherein n is an integer equal to zero, 1, 2 or 3, and R1, R3, R4, R12, R14, R16, R30, R32, R34 and R36 are defined herein. These compounds are useful in the prevention and treatment of infection by HIV and in the prevention, delay in the onset, and treatment of AIDS. The compounds are employed against HIV infection and AIDS as compounds per se or in the form of pharmaceutically acceptable salts. The compounds and their salts can be employed as ingredients in pharmaceutical compositions, optionally in combination with other antivirals, immunomodulators, antibiotics or vaccines.

Description

    FIEND OF THE INVENTION
  • The present invention is directed to tetrahydro-4H-pyrido[1,2-a]pyrimidines, related compounds, and pharmaceutically acceptable salts thereof, their synthesis, and their use as inhibitors of the HIV integrase enzyme. The compounds and pharmaceutically acceptable salts thereof of the present invention are useful for preventing or treating infection by HIV and for treating or delaying the onset of AIDS.
  • BACKGROUND OF THE INVENTION
  • A retrovirus designates human immunodeficiency virus (HIV) is the etiological agent of the complex disease that includes progressive destruction of the immune system (acquired immune deficiency syndrome; AIDS) and degeneration of the central and peripheral nervous system. This virus was previously known as LAV, HTLV-III, or ARV. A common feature of retrovirus replication is the insertion by virally-encoded integrase of proviral DNA into the host cell genome, a required step in HIV replication in human T-lymphoid and monocytoid cells. Integration is believed to be mediated by integrase in three steps: assembly of a stable nucleoprotein complex with viral DNA sequences; cleavage of two nucleotides from the 3' termini of the linear proviral DNA; covalent joining of the recessed 3' OH termini of the proviral DNA at a staggered cut made at the host target site. The fourth step in the process, repair synthesis of the resultant gap, may be accomplished by cellular enzymes.
  • Nucleotide sequencing of HIV shows the presence of a pol gene in one open reading frame [Ratner, L. et al., Nature, 313, 277(1985)]. Amino acid sequence homology provides evidence that the pol sequence encodes reverse transcriptase, integrase and an HIV protease [Toh, H. et al., EMBO J. 4, 1267 (1985); Power, M.D. et al., Science, 231,1567 (1986); Pearl, L.H et al., Nature, 329, 351 (1987)]. All three enzymes have been shown to be essential for the replication of HIV.
  • It is known that some antiviral compounds which act as inhibitors of HIV replication are effective agents in the treatment of AIDS and similar diseases, including reverse transcriptase inhibitors such as azidothymidine (AZT) and efavirenz and protease inhbitors such as indinavir and nelfinavir. The compounds of this invention are inhibitors of HIV integrase and inhibitors of HIV replication. The inhibition of integrase in vitro and HIV replication in cells is a direct result of inhibiting the strand transfer reaction catalyzed by the recombinant integrase in vitro in HIV infected cells. The particular advantage of the present invention is highly specific inhibition of HIV integrase and HIV replication.
  • The following references are of interest as background:
    • US 6380249 , US 6306891 , and US 6262055 disclose 2,4-dioxobutyric acids and acid esters useful as HIV integrase inhibitors.
    • WO 01/00578 discloses 1-(aromatic- or heteroaromatic-substituted)-3-(heteroaromatic substituted)-1,3-propanediones useful as HIV integrase inhibitors.
    • US 2003/0055071 (corresponding to WO 02/30930 ), WO 02/30426 , and WO 02/55079 each disclose certain 8-hydroxy-1,6-naphthyridine-7-carboxamides as HIV integrase inhibitors.
    • WO 02/036734 discloses certain aza- and polyaza-naphthalenyl ketones to be HIV integrase inhibitors.
    • WO 03/016275 discloses certain compounds having integrase inhibitory activity.
    • WO 03/35076 discloses certain 5,6-dihydroxypyrimidine-4-carboxamides as HIV integrase inhibitors, and WO 03/35077 discloses certain N-substituted 5-hydroxy-6-oxo-1,6-dihydropyrimidine-4-carboxamides as HIV integrase inhibitors.
    • WO 03/062204 discloses certain hydroxynaphthyridinone carboxamide that are useful as HIV integrase inhibitors.
    SUMMARY OF THE INVENTION
  • The present invention is directed to novel pyridopyrimidine derivatives and related compounds. These compounds are useful in the inhibition of HIV integrase, the prevention of infection by HIV, the treatment of infection by HIV and in the prevention, treatment, and delay in the onset of AIDS and/or ARC, either as compounds or their pharmaceutically acceptable salts or hydrates (when appropriate), or as pharmaceutical composition ingredients, whether or not in combination with other HIV/AIDS antivirals, anti-infectives, immunomodulators, antibiotics or vaccines.
  • The present invention also includes pharmaceutical compositions containing a compound of the present invention and methods of preparing such pharmaceutical compositions. The present invention further includes the use of the compounds for treating AIDS, delaying the onset of AIDS, preventing AIDS, preventing infection by HIV, and treating infection by HIV.
  • Other embodiments, aspects and features of the present invention are either further described in or will be apparent from the ensuring description, examples and appended claims.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides compounds of Formula II, and pharmaceutically acceptable salts thereof:
    Figure imgb0001
    wherein
    R1 is NR2R5; R2 is CH3; R5 is 1) C(O)CH2SO2CH3, 2) C(O)C(O)N(CH3)2, 3) SO2N(CH3)2, or 4) SO2R20, wherein R20 is:
    Figure imgb0002
    or alternatively R2 and R5 together with the nitrogen atom to which they are attached form
    Figure imgb0003
    or
    Figure imgb0004
    R3 is hydrogen; R4 is p-fluorobenzyl, 4-fluoro-3-methylbenzyl, 3-chlorobenzyl, or 3-chloro-4-methylbenzyl; R12 and R14 are both H, except that when R5 is C(O)C(O)N(CH3)2 and R4 is p-fluorobenzyl and n is 1, then R12 and R14 are either both H or both CH3; and n is an integer equal to 1 or 2.
  • In another feature, R1 is NR2R5; R2 is CH3; R5 is 1) C(O)C(O)N(CH3)2 or 2) SO2R20, wherein R20 is
    Figure imgb0005
    R3 is hydrogen; R4 is p-fluorobenzyl or 4-fluoro-3-methylbenzyl; R12 and R14 are both H, except that when R5 is C(O)C(O)N(CH3)2 and R4 is p-fluorobenzyl and n is 1, then R12 and R14 are either both H or both CH3; and n is an integer equal to 1 or 2.
  • An embodiment of the present invention is a compound, or a pharmaceutically acceptable salt thereof, selected from the group consisting of the compounds set forth in Table 1 below.
  • Other embodiments of the present invention include the following:
    1. (a) A pharmaceutical composition comprising a compound of Formula (II) and a pharmaceutically acceptable carrier.
    2. (b) A pharmaceutical composition which comprises the product prepared by combining (e.g., mixing) an effective amount of a compound of Formula (II) and a pharmaceutically acceptable carrier.
    3. (c) The pharmaceutical composition of (a) or (b), further comprising a therapeutically effective amount of an HIV infection/AIDS treatment agent selected from the group consisting of HIV/AIDS antiviral agents, immunomodulators, and anti-infective agents.
    4. (d) The pharmaceutical composition of (c), wherein the HIV infection/AIDS. treatment agent is an antiviral selected from the group consisting of HIV protease inhibitors, non-nucleoside HIV reverse transcriptase inhibitors, and nucleoside HIV reverse transcriptase inhibitors.
    5. (e) A combination useful for inhibiting HIV integrase, for treating or preventing infection by HIV, or for preventing, treating or delaying the onset of AIDS, which is a therapeutically effective amount of a compound of Formula II and a therapeutically effective amount of an HIV infection/AIDS treatment agent selected from the group consisting of HIV/AIDS antiviral agents, immunomodulators, and anti-infective agents.
    6. (f) The combination of (e), wherein the HIV infection/AIDS treatment agent is an antiviral selected from the group consisting of HIV protease inhibitors, non-nucleoside HIV reverse transcriptase inhibitors and nucleoside HIV reverse transcriptase inhibitors.
  • The present invention also includes a compound of the present invention (i) for use in, (ii) for use as a medicament for, or (iii) for use in the preparation of a medicament for (a) inhibiting HTV integrase, (b) preventing or treating infection by HIV, or (c) preventing, treating or delaying the onset of AIDS. In these uses, the compounds of the present invention can optionally be employed in combination with one or more HIV/AIDS treatment agents selected from HIV/AIDS antiviral agents, anti-infective agents, and immunomodulators.
  • Additional embodiments of the invention include the pharmaceutical compositions, combinations and methods set forth in (a)-(i) above and the uses set forth in the preceding paragraph, wherein the compound of the present invention employed therein is a compound of one of the embodiments, aspects, classes, sub-classes, or features of the compounds described above. In all of these embodiments, the compound may optionally be used in the form of a pharmaceutically acceptable salt.
  • The symbol "
    Figure imgb0006
    " in front of an open bond in the structural formula of a group marks the point of attachment of the group to the rest of the molecule.
  • The compounds of the present invention may have asymmetric centers and may occur, except when specifically noted, as mixtures of stereoisomers or as individual diastereomers, or enantiomers, with all isomeric forms being included in the present invention.
  • The N-substituted hydroxypyrimidinone compounds of the present invention may also occur as tautomers thereof, such as the following tautomer of a compound of Formula II:
    Figure imgb0007
    It is understood that the present invention includes all tautomers of the hydroxypyrimidinone compounds of Formula II, both singly and in mixtures.
  • The compounds of the present inventions are useful in the inhibition of HIV integrase, the prevention or treatment of infection by human immunodeficiency virus (HIV) and the prevention, treatment or the delay in the onset of consequent pathological conditions such as AIDS. Preventing AIDS, treating AIDS, delaying the onset of AIDS, or preventing or treating infection by HIV is defined as including, but not limited to, treatment of a wide range of states of HIV infection: AIDS, ARC (AIDS related complex), both symptomatic and asymptomatic, and actual or potential exposure to HIV. For example, the compounds of this invention are useful in treating infection by HIV after suspected past exposure to HIV by such means as blood transfusion, exchange of body fluids, bites, accidental needle stick, or exposure to patient blood during surgery.
  • The compounds of this invention are useful in the preparation and execution of screening assays for antiviral compounds. For example, the compounds of this invention are useful for isolating enzyme mutants, which are excellent screening tools for more powerful antiviral compounds. Furthermore, the compounds of this invention are useful in establishing or determining the binding site of other antivirals to HIV integrase, e.g., by competitive inhibition. Thus the compounds of this invention are commercial products to be sold for these purposes.
  • Compounds representative of the present invention have been tested for inhibition in an assay for the strand transfer activity of integrase. The assay is conducted in the manner described in WO 02/30930 . The assay is also in accordance with Wolfe, A.L. et al., J. Viral 1996, 70: 1424-1432, for recombinant integrase, except that: (i) the assay uses preassembled integrase strand transfer complexes; (ii) the strand transfer reaction is performed in the presence of inhibitor in 2.5 mM MgCl2 using 0.5 to 5 nM of a 3' FTTC labeled target DNA substrate, and (iii) strand transfer products are detected using an alkaline phosphatase conjugated anti-FITC antibody and a chemiluminescent alkaline phosphatase substrate. Representative compounds of the present invention exhibit inhibition of strand transfer activity in this assay. For example the compounds set forth in Table 1 below were tested in the integrase assay and demonstrated IC50's of about 5 micromolar or less. Further description on conducting the assay using preassembled complexes is found in Hazuda et al., J. Virol. 1997, 71: 7005-7011; Hazuda et al., Drug Design and Discovery 1997, 15: 17-24; and Hazuda et al., Science 2000, 287: 646-650.
  • Certain compounds representative of the present invention have also been tested in an assay for inhibition of acute HIV infection of T-lymphoid cells, conducted in accordance with Vacca, J.P. et al., Proc. Natl. Acad. Sci. USA 1994, 91: 4096. These compounds-including the compounds set forth below in Table 1- demonstrated IC95's of about 20 micromolar or less.
  • The compounds of the present invention can also act as inhibitors of HIV ribonuclease H (RNase H). The human immunodeficiency virus type 1 (HIV-1) reverse transcriptase (RT) catalyzes the conversion of genomic RNA into double-stranded proviral DNA after cell entry, utilizing the RNA- and DNA-dependent polymerase and RNase H activities of the enzyme. HIV-1 RT is an asymmetric dimer consisting of p66 and p51 polypeptides. The catalytic activities of RT are conducted at discrete sites in the p66 subunit; i.e., the N terminus of p66 catalyzes the RNA- and DNA-dependent DNA polymerase activity, an the p15 domain at the C terminus catalyzes RNase H activity. RNase H is required to cleave the RNA strand of the RNA:DNA heteroduplex intermediates in reverse transcription. The compounds of the present invention can selectively bind to and inhibit the RNase H domain of HIV-1 RT. The RNase H inhibition activity of the compounds can be measured using suitable assays known in the art, such as the assay described in Shaw-Reid et al., J. Biol. Chem. 2003, 278 (5): 2777-2780. Accordingly, the present invention includes a method of inhibiting HIV RNase H in a subject in need of such inhibition which comprises administering to the subject an effective amount of a compound of the invention. The present invention further includes a compound of the present invention (i) for use in, (ii) for use as a medicament for, or (iii) for use in the preparation of a medicament for inhibiting HIV RNase H.
  • The compounds of the present invention may be administered in the form of pharmaceutically acceptable salts. The term "pharmaceutically acceptable salt" refers to a salt which possesses the effectiveness of the parent compound and which is not biologically or otherwise undesirable (e.g., is neither toxic nor otherwise deleterious to the recipient thereof). Suitable salts include acid addition salts which may, for example, be formed by mixing a solution of the compound of the present invention with a solution of a pharmaceutically acceptable acid such as hydrochloric acid, sulfuric acid, acetic acid, trifluoroacetic acid, or benzoic acid When the compounds of the invention carry an acidic moiety, suitable pharmaceutically acceptable salts thereof can include alkali metal salts (e.g., sodium or potassium salts), alkaline earth metal salts (e.g., calcium or magnesium salts); and salts formed with suitable organic ligands such as quaternary ammonium salts. Also, in the case of an acid (-COOH) or alcohol group being present, pharmaceutically acceptable esters can be employed to modify the solubility or hydrolysis characteristics of the compound.
  • For the purpose of inhibiting HIV integrase or HIV RNase H, preventing or treating HIV infection or preventing, treating or delaying the onset of AIDS, the compounds of the present invention may be administered orally, parenterally (including subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques), by inhalation spray, or rectally, in the form of a unit dosage of a pharmaceutical composition containing a therapeutically effective amount bf the compound and conventional non-toxic pharmaceutically-acceptable carriers, adjuvants and vehicles.
  • The term "administration" and variants thereof (e.g., "administering" a compound) in reference to a compound of the invention mean providing the compound or a prodrug of the compound to the individual in need of treatment. When a compound of the invention or a prodrug thereof is provided in combination with one or more other active agents (e.g., antiviral agents useful for treating HIV infection or AIDS), "administration" and its variants are each understood to include concurrent and sequential provision of the compound or prodrug and other agents.
  • As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combining the specified ingredients in the specified amounts.
  • By "pharmaceutically acceptable" is meant that the ingredients of the pharmaceutical composition must be compatible with each other and not deleterious to the recipient thereof.
  • The term "subject" (alternatively referred to herein as "patient") as used herein refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
  • The term "therapeutically effective amount" as used herein means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation or prophylaxis of the symptoms of the disease or condition being treated or prevented. The term also includes herein the amount of active compound sufficient to inhibit HIV integrase and/or RNase H and thereby elicit the response being sought. When the active compound (i.e., active ingredient) is administered as the salt, references to the amount of active ingredient are to the free acid or free base form of the compound.
  • The pharmaceutical compositions may be in the form of orally-administrable suspension or tablets or capsules, nasal sprays, sterile injectible preparations, for example, as sterile injectible aqueous or oleagenous suspensions or suppositories. These compositions can be prepared by methods and contain excipients which are well known in the art. Suitable methods and ingredients are described in Remington's Pharmaceutical Sciences, 18th edition, edited by A. R. Gennaro, Mack Publishing Co.,1990, which is herein incorporated by reference in its entirety.
  • The compounds of this invention can be administered orally in a dosage range of 0.001 to 1000 mg/kg of mammal (e.g., human) body weight per day in a single dose or in divided doses. One referred dosage range is 0.01 to 500 mg/kg body weight per day orally in a single dose or in divided doses. Another preferred dosage range is 0.1 to 100 mg/kg body weight per day orally in single or divided doses. For oral administration, the compositions can be provided in the form of tablets or capsules containing 1.0 to 500 milligrams of the active ingredient, particularly 1, 5,10,15, 20, 25, 50, 75, 100, 150, 200, 250, 300, 400, and 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. The specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
  • As noted above, the present invention is also directed to use of the HIV integrase inhibitor compounds of the present invention with one or more agents useful in the treatment of HIV infection or AIDS. For example, the compounds of this invention may be effectively administered, whether at periods of pre-exposure and/or post-exposure, in combination with effective amounts of one or more HIV/AIDS antivirals, imunomodulators, antiinfectives, or vaccines useful for treating HIV infection or AIDS, such as those disclosed in Table 1 of WO 01/38332 or in the Table in WO 02/30930 , both documents being herein incorporated by reference in their entireties. It will be understood that the scope of combinations of the compounds of this invention with HIV/AIDS antivirals, immunomodulators, anti-infectives or vaccines is not limited to the list in the above-referenced Tables in WO 01/38332 and WO 02/30930 , but includes in principle any combination with any pharmaceutical composition useful for the treatment of AIDS. The HIV/AIDS antivirals and other agents will typically be employed in these combinations in their conventional dosage ranges and regimens as reported in the art, including, for example, the dosages described in the physicians' Desk Reference, 57th edition, Thomson PDR, 2003. The dosage ranges for a compound of the invention in these combinations are the same as those set forth above.
  • Abbreviations used in the instant specification, particularly the Schemes and Examples, include the following: AIDS = acquired immunodeficiency syndrome, ARC = AIDS related complex, Bn = benzyl, CBZ (or Cbz) = benzyloaycarbonyl, DBU =1,8-diazabicyclo[5.4.0]undec-7-ene, DMAD = dimethylacetylenedicarboxylate, DMF = N,N-dimethylformamide, DMSO = dimethylsulfoxide, EtOAc = ethyl acetate, FIA-MS = flow injection analysis mass spectrometry, h = hour(s), HIV = human immunodeficiency virus, HPLC = high performance liquid chromatography, IPA = isopropanol, LDA = lithium diisopropylamide, Me = methyl, MeOH = methanol, NMP = N-methyl pyrrolidinone, NMR = nuclear magnetic resonance, Pd/C = palladium on carbon catalyst, RP-HPLC = reversed phase HPLC, TFA = trifluoroacetic acid, THF = tetrahydrofuran.
  • The compounds of the present invention can be readily prepared according to the following reaction schemes and examples, or modifications thereof, using readily available starting materials and reagents. In the reactions shown below, it is also possible to make use of variants which are themselves known to those of ordinary skill in this art, but are not mentioned in greater detail. Furthermore, other methods for preparing compounds of the invention (including compounds embraced by Formula A and Formula I but whose preparation is not literally described below) will be readily apparent to the person of ordinary skill in the art in light of the following reaction schemes and examples. Unless otherwise defined, the variables listed in Schemes 1, A, B, C, D and E have the following meanings:
  • P^ is hydrogen or a protective group, e.g., an ester such as, but not limited to, benzoate and pivalate, or an ether such as, but not limited to, a benzyl ether, that is normally removed under the conditions employed to convert the methyl ester to the amide or is removed in a different step. The protective group is typically used for synthetic and/or purification reasons.
    • R^ is hydrogen or C1-6 alkyl.
    • Y is hydrogen or NRsaRsb.
    • Rsa is as defined for R5 or Rsa and Rsb, together with the nitrogen atom to which they are attached, form a heterocyclic ring containing 1 or 2 heteroatoms as defined for Formula II.
    • Rsb is as defined for R2.
    • Rs1 is hydrogen.
    • Rs2 is as defined for R4.
  • The compounds of the present invention can be prepared by coupling the appropriate amines with suitable substituted alkyl 3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H pyrido[1,2-a] pyrimidine -2-carboxylate (or carboxylic acids or halides) or alkyl 3-hydroxy-4-oxo-4,6,7,8-tetrahydropyrrolo[1,2-a]pyrimidine-2-carboxylate (or carboxylic acids or halides) or alkyl 3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxylate (or carboxylic acids or halides) or alkyl 3-hydroxy-4-oxo-6,7,8,9,10,11-hexahydro-4H-pyrimido[1,2-a]azepine-2-carboxylate (or carboxylic acids or halides), as represented by Scheme 1.
    Figure imgb0008
  • Methods for coupling carboxylic acid derivatives with amines to form carboxamides are well known in the art. Suitable methods are described, for example, in Jerry March, Advanced Organic Chemistry, 3rd edition, John Wiley & Sons, 1985, pp. 370-376. Amines of formula 1-2 can be prepared using the methods described in Richard Larock, Comprehensive Organic Transformations. VCH Publishers Inc,1989, pp 385-438, or routine variations thereof.
  • Scheme A depicts a general synthesis of carboxamides A-5. The methyl ester A-4 can be reacted with an amine 1-2 in solvents like DMF, methanol, ethanol, toluene, NMP at the appropriate temperature (e.g., from 20 to 150°C) to give the final compound A-5. Methyl ester A-4 can be synthesized by one of three synthetic routes. In the first route, amidine hydrochloride A-1a (sX=H; -sY=H) can be reacted with dimethyl 2-(benzyloxy)-3-oxosuccinate in the presence of a base to afford the protected methyl ester intermediate A-2, which can be readily deprotected to afford the methyl ester A-4. In a second route, amidoxine A-1b (sX=OH; sY=H), obtained in three steps from tert-butyl benzyloxycarbamate, can be reacted with DMAD to afford the cyclic intermediate A-3a, which can be rearranged by heating in an appropriate solvent (e.g., xylene) to afford the methyl ester A-5. In a third route, amidoxyme A-1c (sX=H;sY=OH) can be reacted with DMAD in an appropriate solvent (e.g., acetonitrile) to afford the intermediate A3-b, which can be rearranged to the methyl ester A-4 by heating in an appropriate solvent (e.g., xylene). All three of these routes can be applied to amidines and amidoxymes containing ring substituents. Scheme A is exemplified in Reference Examples 1 and 9.
    Figure imgb0009
  • Scheme B shows a method for preparing compounds of the present invention that contain an amine, ether, thioether, sulfoxide or sulfone group at the 9-position of the 3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide core. The bromo derivative B-1 can be obtained from methyl ester A-4 by first protecting the hydroxy group on A-4 with a suitable protective group (e.g., conversion to a benzoate or pivalate or to benzyloxy) and then contacting the protected A-4 with a brominating agent (e.g., NBS). The bromo derivative B-1 can then be treated with a nucleophile ("Nu"; e.g. an amine, thiol or alcoholate) to afford with or without isolation the methyl ester intermediate B-2, that is reacted with the desired amine to give the final product B-3. If the nucleophile is a thiol or contains an oxidizable sulfur, an oxidation step to obtain the sulfoxide or sulfone can be included in the scheme. If the nucleophile contains an ester, the ester can be converted to an amide by routine chemistry after the synthesis of B-3. Scheme B is exemplified in Example 2.
    Figure imgb0010
  • Scheme C depicts a general synthesis of derivatives C-3 or C-4 containing an aliphatic ring substituent such as amide, sulfonamide, sulfonylurea, carbamate, or urea. The bromo derivative B-1 can be treated with benzylamine C-1 and then hydrogenated or reacted directly with amine C-1a to give intermediate C-2, which can then be treated with amine 1-2, with or without isolation, and then coupled with a carboxylic acid or reacted with a carbonyl chloride (or sulfonyl chloride or sulfamoyl chloride) or isocyanate to afford the final product C-3. Scheme C is exemplified in Examples 3,4,10 and 11. The last two steps can be reversed.
    Figure imgb0011
  • Scheme D shows the synthesis of homochiral compounds C-3, C3a,b, and C-4. The bromo derivative B-1 is displaced with chiral amines D-1 to give a mixture of diastereoisomers, with subsequent or simultaneous removal of the protecting group. The amino group at the 9-position is reductively alkylated with aldehydes or ketones D-2 to afford the intermediate D-3. The mixture of diastereoisomers can be separated by crystallization or chromatography to give the single diastereoisomers D-3a,b. Rs6 can be removed by hydrogenation to afford the homochiral C-2a,b intermediate. A subsequent reaction with amine 1-2 and coupling with carboxylic acid or treatment with carbonyl chloride (or sulfonyl chloride or sulfamoyl chloride) or isocyanate affords the final homochiral product C-3a,b. An additional step can be performed to produce homochiral C-4 compounds. Scheme D is exemplified in Examples 5, 6, 7, 8 and 12.
    Figure imgb0012
  • The following examples serve only to illustrate the invention and its practice.
  • EXAMPLE 1 (Reference) N-(4-Fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide
  • Figure imgb0013
  • Step 1a: Tert-butyl benzyloxy(4-cyanobutyl)carbamate (Bergeron, R. J., McManis, J. S., Tetrahedron 45 (16), 4939-4944 (1989).
  • To a solution of tert-butyl benzyloxycarbamate in anhydrous dimethylformamide were added 5 mol% of sodium iodide and portionwise 1.36 eq. of sodium hydride (60 % dispersion in mineral oil). The mixture was stirred at room temperature for 15 min. before 1.05 eq. of 4-chlorovaleronitrile were added. The mixture was heated to 85°C and stirred for 3.5 h. After cooling to room temperature the mixture was quenched with water and extracted with diethyl ether. The combined organic phases were concentrated and washed with half saturated aq. sodium thiosulfate, water and brine. The organic phase was dried over sodium sulfate, filtered and concentrated under reduced pressure. The oily residue was washed with petroleum ether and dried under high vacuum to yield Tert-butyl benzyloxy(4-cyanobutyl)carbamate as a light yellow oil.
    1H-NMR (400 MHz, CDCl3) δ: 7.38 (m, 5H). 4.84 (s, 2H), 3.45 (t, J = 6.4 Hz, 2H), 2.34 (t, J = 6.8 Hz, 2H), 1.70 (m, 4H), 1.52 (s, 9H). MS m/z: 271 (M+H)+.
  • Step 2a: 1-(Benzyloxy)piperidin-2-imine hydrochloride
  • Tert-butyl benzyloxy(4-cyanobutyl)carbamate was dissolved in a solution of 4 M HCl in 1,4-dioxane and the mixture was stirred for 18 h at room temperature. The solvent was removed under reduced pressure and the residue was treated with ethyl acetate and diethyl ether. A solid formed which was washed with diethyl ether, filtered and dried under high vacuum to give 1-(benzyloxy)piperidin-2-imine hydrochloride as a pale yellow solid.
    1H-NMR (400 MHz, DMSO-d6) δ 9.53 (s, 1H). 8.97 (s, 1H), 7.57-7.41 (m, 5H), 5.05 (s, 2H), 3.67 (t, J=6.0 Hz, 2H). 2.64 (t, J=6.4 Hz, 2H), 1.90-1.84 (m, 2H), 1.69-1.63 (m, 2H). MS m/z: 205 (M+H)+.
  • Step 3a: 2-Iminopipendin-1-ol hydrochloride
  • A solution of 1-(benzyloxy)piperidin-2-imine hydrochloride in methanol, containing palladium on charcoal (10 %, w/w) was stirred under hydrogen at atmospheric pressure for 3 h. The catalyst was filtered off and the solution was concentrated to dryness under reduced pressure. The residue was triturated with diethyl ether, filtered and dried under high vacuum to afford 2-iminopiperidin-1-ol hydrochloride as a pale yellow solid. 1H-NMR (400 MHz, DMSO-d6) δ: 11.76 (s, 1H), 8.82 (s, 1H). 8.49 (s, 1H), 3.63 (t, J=6.0 Hz, 2H), 2.63 (t, J=6.0 Hz, 2H), 1.87 (m. 2H), 1.66 (m, 2H). 13C-NMR (150 MHz, DMSO-d6) δ: 159.06, 50.92, 25.76, 22.01,1722.
    MS m/z: 115 (M+H)+.
  • Step 4a: Methyl 2-(2-methoxy-2-oxoethyl)-56,7,8-tetrahydro-2H-[1,2,4]oxadiazolo[2,3-a]pyridine-2-carboxylate
  • To a solution of 2-iminopiperidin-1-ol hydrochloride in chloroform was added triethylamine. The mixture was stirred for 5 min. at room temperature, then cooled to 0 °C and 1.2 eq. of dimethyl acetylenedicarboxylate were added dropwise under stirring. The cooling bath was removed and the mixture was stirred at room temperature for one hour. The solvent was removed under reduced pressure and the solution was partitioned between ethyl acetate and half saturated aq. ammonium chloride. The aqueous phase was further extracted with ethyl acetate. The combined organic phases were dried over sodium sulfate and filtered through silica gel. The solvent was removed under vacuum to afford methyl 2-(2-methoxy-2-oxoethyl)-5,6,7,8-tetrahydro-2H-[1,2,4]oxadiazolo[2,3-a]pyridine-2-carboxylate as a light yellow oil.
    1H-NMR (400 MHz, CDCl3) δ: 3.82 (s, 3H), 3.70 (s, 3H), 3.51 (m 1H), 3.36 (m, 1H), 3.31 (d, J =16.6 Hz, 1H), 2.98 (d, J = 16.6 Hz, 1H), 2.53 (m, 2H), 1.94 (m, 2H), 1.74 (m, 2H). 13C-NMR (100 MHz, CDCl3) δ: 169.15, 168.88, 1b4.97, 103.27, 55.71, 52.97, 51.84, 42.26, 26.06, 23.49, 22.83.
    MS m/z: 257 (M+H)+.
  • Step 5a: Methyl 3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate
  • A solution of methyl 2-(2-methoxy-2-oxoethyl)-5,6,7,8-tetrahydro-2H-[1,2,4]oxadiazolo [2,3-a]pyridine-2-carboxylate in anhydrous o-xylene was placed in a double necked round bottom flask. The flask was equipped was a thermometer and closed with a septum. The mixture was heated to 148-150 °C for 5 h. The heating bath was removed and the mixture was left standing at room temperature for 16 h. To the mixture containing a precipitate was added diethyl ether. After 5 min. the precipitate was filtered off, washed with diethyl ether and dried under vacuum. Product methyl 3-hydroay-4-oxo-6,7,8,9-tetrahydro-4H pyrido[1,2-a]pyrimidine-2-carboxylate was obtained as a pale brown solid.
    1H-NMR (400 MHz, DMSO-d6) δ: 10.03 (s, 1H), 3.86 (t, J = 6.0 Hz, 2H), 3.80 (s, 3H), 2.75 (t, J = 6.8 Hz, 2H), 1.90-1.70 (m, 4H). 13C-NMR (150 MHz, DMSO-d6) δ:165.81, 158.65, 148.60, 143.10, 127-07, 51.98, 42.87, 30.32, 20.91,18.40.
    MS m/z: 225 (M+H)+.
  • The following procedure is an alternative route for the synthesis of methyl 3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate
  • Step 1b: Dimethyl 2-(benzyloxy)-3-oxosuccinate
  • A solution of methyl(benzyloxy)acetate (1 eq) and dimethyl oxalate (1.2 eq) in dry THF was cooled to -78°C and LDA (2M in THF-heptane, 1.2 eq) was added dropwise. After stirring for an hour the cold bath was removed, and stirring was continued for an additional hour. The reaction was quenched at 0°C by pouring into cold 1N aq HCl, and the aqueous phase was extracted with EtOAc; the organic layer was washed with brine, dried and concentrated to give a crude that was used without further purification.
  • Step 2b: Methyl 3-(benzyloxy)4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate
  • Commercially available 2-iminopiperidine hydrochloride (1.5 eq) was added at room temperature to a solution of oxosuccinate prepared in Step 1b (1 eq) in MeOH. After dropwise addition of neat DBU (4.5 eq), the reaction mixture was stirred for 2 days. Evaporation of the solvent gave a residue that was taken into EtOAc and washed with 1N HCl and brine; the organic layer was dried on Na2SO4 and the solvent removed. The crude was used without further purification.
    An analytical sample of this product was purified by flash chromatography (Petroleum ether /EtOAc 1:2 to 1:5), and has the following spectroscopical data:
    1H-NMR (400 MHz, CDCl3) δ: 7.49-7.30 (m, 5H). 5.25 (s, 2H), 4.00 (t, J=6.2 Hz, 2H), 3.86 (s, 3H), 2.94 (t, J=6.6 Hz, 2H), 2.01-1.95 (m, 2H), 1.92-1.87 (m, 2H).
    13C-NMR (75 MHz, CDCl3) δ: 164.1, 159.3, 154.1, 141.1, 140.6, 136.0, 128.1, 127.8, 127.7, 73.8, 52.2, 42.7, 30.9, 21.0, 18.4.
    MS m/z: 315 (M+H)+.
  • Step 3b: Methyl 3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate
  • Intermediate methyl 3-(benzyloxy)-4-oxo-6,7,8,9-tetrahydro-4H-pyxido[1,2a]pyrimidine-2-carboxylate prepared in Step 2b was dissolved in MeOH and catalytic 10% Pd/C was added at room temperature. The mixture was stirred under H2 atmosphere for 3.5 hours. Filtration of the catalyst and evaporation of methanol gave a residue to which diethyl ether was added; filtration afforded methyl 3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate as a pale brown solid with the spectroscopical properties identical to the compound described in Step 5a.
  • Step 6: N-(4-Fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide .
  • A solution of methyl 3-hydroxy4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate obtained in Step 3b or Step 5a and 4-fluoro-benzylamine (2 eq.) in methanol was stirred and heated to 65°C for 22 h. The solvent was removed under reduced pressure and the title product was obtained by preparative RP-HPLC, using water (0.1 % TFA) and acetonitrile (0.1 % TFA) as eluents (column: C18). The pooled product fractions were lyophilized to afford the title compound as a fluffy white material.
    1H-NMR (400 MHz, DMSO-d6) δ: 12.12 (s, 1H). 9.35 (m, 1H), 7.36 (m, 2H), 7.15 (m, 2H), 4.44 (m, 2H), 3.84 (t, J=6.4Hz, 2H), 2.80 (t, J=6.8Hz, 2H), 1.90-1.73 (m, 4H). MS m/z: 318 (M+H)+.
  • EXAMPLE 2 N-(4-Fluorobenzyl)-3-hydroxy-9-morpholin-4-yl-4-oxo-6,7,8,9-tecrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide hydrochloride
  • Figure imgb0014
  • Step 1: Methyl 3-(benzoyloxy)-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyiimidine-2-carboxylate
  • To a solution of methyl 3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate (obtained following Example 1) in pyridine was added benzoic anhydride (1.55 eq.). The mixture was stirred at room temperature for 16 h. The solvent was removed under reduced pressure and the residue was partitioned between ethyl acetate and 0.5 M aq. HCl. The aqueous phase was extracted with ethyl acetate and the combined organic phases were washed with 0.5 M aq. HCl, water and brine. The organic phase was dried over sodium sulfate, filtered and concentrated to dryness under vacuum. Title compounds was obtained after flash chromatography (eluent petroleum ether/ethyl acetate, 1:2) as a colorless solid.
    1H-NMR (400 MHz, DMSO-d6) δ: 8.07 (m, 2H), 7.78 (m, 1H). 7.62 (m, 2H), 3.86 (t, J = 6.0 Hz, 2H), 3.74 (s, 3H), 2.92 (t, J = 6.4 Hz, 2H), 1.93-1.81 (m, 4H). MS m/z: 329 (M+H)+.
  • Step 2: Methyl 3-(benzoyloxy)-9-bromo-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate
  • A mixture of methyl 3-(benzoyloxy)-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate , N-bromo-succinimide (1.2 eq.) and dibenzoylperoxide (70 %, 0.13 eq.) in carbon tetrachloride was stirred under reflux for one hour. The mixture was cooled to room temperature, the succinimide was filtered off and the solvent was removed under reduced pressure. Methyl 3-(benzoyloxy)-9-bromo-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate was obtained after flash chromatography (eluent petroleum ether/ethyl acetate, 1:1) as a pale yellow oil.
    1H NMR (400 MHz, DMSO-d6) δ: 8.08 (m, 2H), 7.79 (m, 1H), 7.63 (m, 2H), 5.58 (m, 1H), 4.24 (m, 1H), 3.77 (s, 3H), 3.72 (m, 1H), 2.43-2.35 (m, 1H), 2.30-2.05 (m, 3H). MS m/z: 409/407
    (M+H)+.
  • Step 3: N-(4-Fluorobenzyl)-3-hydroxy-9-morpholin-4-yl-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamido.
  • To a solution of methyl 3-(benzoyloxy)-9-bromo-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboaylate in DMF was added morpholine (3 eq.) and the mixture was stirred at room temperature for 1h. The solvent was removed under reduced pressure and the residue was triturated with diethyl ether. The crude material was dissolved in methanol, 4-fluoro-benzylamine (3 eq.) was added and the mixture was stirred for 1.5 h at 65°C. The solvent was removed under reduced pressure and the product was purified by preparative RP-HPLC, using water (0.1 % TFA) and acetonitrile (0.1 % TFA) as eluents (column: C18). The pooled product fractions were lyophilized and redissolved in 1N HCl. The solvent was removed under reduced pressure and the residue was lyophilized from water/ acetonitrile to afford the hydrochloride salt of N-(4-fluorobenzyl)-3-hydroxy-9-morpholin-4-yl-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide as a slightly pink fluffy material.
    1H-NMR (400 MHz, DMSO-d6) δ: 12.34 (s,1H), 10.99 (s, 1H), 10.47 (s, 1H), 7.44 (m; 2H), 7.16 (m. 2H), 4.85 (m, 1H),4.60-4.40 (m, 3H), 4.10-3.85 (m, 4H), 3.60-3.05 (m, 5H obscured by water signal), 2.35-2.15 (m, 2H), 2.03-1.80 (m, 2H). MS m/z: 403 (M+M)+.
  • EXAMPLE3 3 (+/-)-9-[[(Dimethylamino)sulfonyl](methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide C-3
  • Figure imgb0015
  • Step 1: Methyl 9-[benzyl(methyl)amino]-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate hydrochloride
  • To a stirred solution of the bromo derivative methyl 3-(benzoyloxy)-9-bromo-4-oxo-6,7,8,9-tetrahydto-4H-pyrido[1,2-a]pyrimidine-2-carboxylate (obtained in Example 2, Step 2) in anhydrous dimethylformamide was added N-benzyl-N-methylamine (3 eq.). The mixture was stirred for 1.5 h at room temperature before diethyl ether and 2 M HCl in diethyl ether were added. The mixture was stirred for 5 min., the formed precipitate was filtered off and washed with diethyl ether. The precipitate was dissolved in anhydrous methanol and the solution was concentrated to dryness under reduced pressure. The title crude product obtained as a pale yellow oil, which contained an excess of N-benzyl-N-methylamine hydrochloride, was used without further purification.
    MS m/z: 344 (M+H)+.
  • Step 2: Methyl 3-hydroxy-9-(methylamino)-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate
  • A solution of crude obtained in Step 1 in methanol, containing palladium on charcoal (10 % w/w) was stirred under hydrogen at atmospheric pressure for 3 h. The catalyst was filtered off and the solution was concentrated to dryness under reduced pressure. The residue was triturated with diethyl ether, filtered and dried under high vacuum to afford the crude product as a yellow solid, which was used in the next step without further purification.
    MS m/z: 254 (M+H)+.
  • Step 3: N-(4-Fluombenzyl)-3-hydroxy-9-(methylamino)-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide
  • To a solution of the crude obtained in Step 2 in dry methanol were added an excess of triethylamine. The solvent was removed under reduced pressure and then under high vacuum. The oily residue was dissolved in anhydrous methanol and 4-fluoro-benzylamine was added (3.1 eq. th.). The mixture was stirred and heated to 60 °C overnight. After cooling to room temperature the solvent was removed under reduced pressure. The residue was triturated with diethyl ether and left under high vacuum for 15 min. The title crude product was obtained as a yellow solid, which contained an excess of 4-fluoro-benzylamine (ca. 3.5 eq.) and was used without further purification.
    MS m/z: 347 (M+M)+.
  • Step 4: (+/-)-9-[[(Dimemylamino)sulfonyl](methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide
  • To a solution of crude compound obtained in Step 3 in a 2:1 mixture of tetrahydrofuran and 2 M aq. sodium hydroxide was added N,N-dimethylsulfamoyl chloride (4.6 eq.). The mixture was stirred at roomtemperature for 16 h. The mixture was concentrated under reduced pressure and the product was isolated by preparative RP-HPLC, using water (0.1 % TFA) and acetonitrile (0.1 % TFA) as eluents (column: C18). The pooled product fractions were lyophilized to afford the title compound as a fluffy, slightly pink material.
    1H-NMR (300 MHz, CDCl3) δ: 11.95 (s, 1H), 9.13 (m, 1H), 7.38 (m, 2H), 7.04 (m, 2H), 4.98 (m, 1H), 4.56 (m, 2H), 4.36 (m, 1H), 3.62 (m, 1H), 2.84 (s, 6H), 2.57 (s, 3H), 2.38-1.85 (m, 4H). 13C-NMR (100 MHz, CDCl3) δ: 167.53, 162.55, 160.11, 157.74, 145.76 144.11, 132.70, 128.89, 128.81, 124.82, 114.60, 114.39, 58:06, 42.93, 41.53, 37.00, 29.03, 23.89, 20.09.
    MS m/z: 454 (M+H)+.
  • EXAMPLE 4 (+/-)-N 1-(2-{[(4-Fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N 1,N 2,N 2-trimthylethanediamide
  • Figure imgb0016
  • Step 1: (+/-)-N 1-(2-{[(4-Fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N 1,N 2,N 2-trimethylethanediamide
  • To a stirred solution of crude N-(4-fluorobenzyl)-3-hydroxy-9-(methylamino)-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide (synthesized as described in Example 3, Step 3) in dichloromethane were added 6 eq. of triethylamine and 6 eq. of methyl chlorooxoacetate. The mixture was stirred at room temperature for 2 h, the solvent was removed under reduced pressure and the residue was dissolved in a solution of dimethylamine (2 M) in tetrahydrofuran. The mixture was stirred at 57°C overnight. After cooling to room temperature, the solvent was removed under reduced pressure and the product was isolated by preparative RP-HPLC, using water (0.1 % TPA) and acetonitrile (0.1 % TFA) as eluents (column: C18). The pooled product fractions were lyophilized to afford the title compound as a fluffy, slightly pink material. The product was a mixture of rotamers by NMR.
    1H-NMR (400 MHz, DMSO-d6) δ: 12.05 (s, 0.2H), 11.89 (s, 0.8H), 9.21 (m, 0.8H), 8.74 (m, 0.2H), 7.40-7.28 (m, 2H), 7.20-7.10 (m, 2H), 5.17 (m, 0.8H), 4.63-4.35 (m, 2.2H), 4.13-4.00 (m, 1H), 3.65-3.53 (m, in part overlaid by water signal), 2.95-2.75 (m, 9H), 2.15-1.80 (m, 4H). 13C-NMR (100 MHz, DMSO-d6) δ:167.87, 167.73, 165.92, 165.46, 164.51, 164.30, 162.42, 160.01. 157.50,157.41,146.27,146.18,145.76,145.49,134.44,129.43,129.35,129.08,129.00,125.17, 125.05,115.07,114.85, 57.47, 53.60, 43.14, 41.37, 36.49, 35.9S, 32.92, 32.64, 32.36, 28.19, 23.88, 22.12, 19.67,19.35.
    MS m/z: 446 (M+H)+
  • EXAMPLE 5 (+)-N 1-(2-{[(4-Fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pirido[1,2-a]pyrimidin-9-yl)-N 1,N 2,N 2-trimethylethanediamide
  • Figure imgb0017
  • Step 1: (+)3-Hydroxy-2-(methoxycarbonyl)-N-methyl-4-oxo-N-[(1S)-1-phenylethyl]-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-ammonium trifluoroacetate
  • To a 7:3 mixture (v/v) of methanol and water at -30°C, containing (1S)-1-phenylethylamine (4.5 eq.) was added bromo derivative methyl 3-(benzoyloxy)-9-bromo4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxylate (synthesized as reported in Example 2, Step 2) (1.0 eq.). The mixture was stirred vigorously for 1.5 h at -30 °C. The cooling bath was removed and stirring was continued for 1 h at room temperature. The pH was adjusted to ca. 5 with acetic acid before 37 % aqueous formaldehyde (11.5 eq.) and sodium cyanoborohydride (3.25 eq.) were added. After stirring at room temperature for 20 min. the volume was reduced to ca. 1/4 under reduced pressure. A formed white precipitate was filtered off and the filtrate was acidified to pH 2-3 with trifluoroacetic acid. The solution was applied on cation-exchange resin cartridges (Varian MEGA BOND ELUTE SCX), the cartridges washed with methanol and the crude product was eluted with 2 M ammonia in methanol. The pooled eluents were concentrated to dryness under reduced pressure and the oily residue was dissolved in methanol and neutralized with trifluoroacetic acid. After removal of the solvent an oily residue was obtained The resulting diasteroisomers in 1:3 ratio were separated by preparative RP-HPLC-purification (column: C18) eluents water (0.1 % TFA), acetonitrile (0.1 % TFA). The major diasteroisomer was eluted as second peak and after lyophilization the title compound was obtained as a slightly pink solid.
    1H-NMR (500 MHz, pyridine-d5) δ: 7.53 (m, 2H), 7.39 (m, 2H), 7.29 (m, 1H), 4.45 (m,1H), 4.38 (m, 1H), 4.14 (m, 1H), 3.91 (s, 3H), 3.80 (m,1H), 2.13 (s, 3H), 1.95-1.82 (m, 3H), 1.70-1.60 (m, 1H), 1.49 (d, J=6.4 Hz, 3H).
    MS m/z: 358 (M+H)+.
  • Step 2: (-)-3-Hydroxy-2-(methoxycarbonyl)-N-methyl-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-ammonium trifluoroacetate
  • A solution of (+)3-hydroxy-2-(methoxycarbonyl)-N-methyl-4-oxo-N-[(1S)-1-phenylethyl]-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-ammonium trifluoroacetate in methanol, containing palladium on charcoal (10 %, w/w) was stirred under hydrogen at atmospheric pressure for 1.5 h. The catalyst was filtered off and the solution was concentrated to dryness under reduced pressure to afford the title compound as a slightly pink oil.
    1H-NMR (400 MHz, CD3OD) δ: 4.41 (m, 1H), 4.14 (m, 1H), 3.99 (s, 3H), 3.91 (m,1H), 2.86 (s, 3H), 2.50 (m, 1H), 2.26 (m, 1H), 2.08 (m, 1H), 1.86 (m, 1H).
    MS m/z: 254 (M+H)+.
  • Step 3: (+)-N 1-(2-{[(4-Fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N 1,N 2,N 2-trimethylethanediamide
  • A solution of (-)-3-Hydroxy-2-(methoxycarbonyl)-N-methyl-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-amnonium trifluoroacetate, p-fluoro-benzylamine (2.2 eq.) and triethylamine (1.3 eq.) in methanol was stirred and heated to 65°C for 3 h. The solvent was removed under vacuum and the residue was dissolved in anhydrous dichloromethane. Methyl chlorooxoacetate (5 eq.) and triethylamine (5 eq.) were added and the mixture was stirred at room temperature for 50 min. The solvent was removed under reduced pressure and the residue was dissolved in a 2 M solution of dimethylamine in tetrahydrofuran. The mixture was stirred at 57°C overnight. After cooling to room temperature the solvent was removed under reduced pressure and the product was isolated by preparative RP-HPLC, using water (0.1 % TFA) and acetonitrile (0.1 % TFA) as eluents (Column: C18). The pooled product fractions were lyophilized to afford the title product as a fluffy white material (ee 94.4 %).
  • The compound was dissolved in ethylacetate/heptane (3:2.5 (v/v) mixture) and left standing at room temperature for four days. The supernatant from the formed precipitate was taken off, concentrated under reduced pressure and the residue was lyophilized from water/acetonitrile to afford enantiopure title product e.e.100 %(e.e. determined by Chiral HPLC Chiralpak AS, mobile phase 0.2% TPA n-Hex/IPA) with spectroscopical properties identical to the compound described in Example 4, Step 1.
    [α]20 D= + 36.5±2.5° (C = 0.63 in ethanol).
  • EXAMPLE 6 (-)-N 1-(2-{[(4-Fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)N 1,N 2,N 2-trimethylethanediamide
  • Figure imgb0018
  • Step 1 : (-)-3-Hydroxy-2-(methoxycarbonyl)-N-methyl-4-oxo-N-[(1R)-1-phenylethyl]-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-ammonium trifluoroacetate
  • Title compound was obtained using (1R)-1-phenylethylamine and following the procedure describe in Example 5, Step 1.
    1H-NMR (400 MHz, pyridine-d5) δ: 7.55 (m, 2H), 7,40 (m, 2H), 7.29 (m, 1H), 4.47 (m, 1H), 4.39 (m, 1H), 4.15 (m, 1H), 3.92 (s, 3H), 3.80 (m, 1H), 2.14 (s, 3H), 1.95-1.82 (m, 3H), 1.72-1.60 (m, 1H), 1.50 (d, J=6.4 Hz, 3H). MS m/z: 358 (M+H)+. [α]20 D = - 15.1° (C = 0.55 in methanol).
  • Step 2: (+)-3-Hydroxy-2-(methoxycarbonyl)-N-methyl-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-ammonium trifluoroacetate
  • Title compound was synthesized following the procedure described in Example 5, Step 2, using as starting material the compound synthesized in the previous Step 1. It showed identical spectroscopical properties, except for the optical rotation.
    [α]20 D = +18.7° (C = 0.41 in methanol).
  • Step 3: (-)-N 1-(2-{[(4-Fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N 1,N 2,N 2-trimethylethanediamide
  • The compound was synthesized following the same procedure described in Example 5, Step 3 using the compound prepared in the previous Step 2. The title compound (e.e. 93 %) was dissolved in ethanol and the formed precipitate was filtered off. The obtained solution was concentrated to dryness under vacuum and the residue was redissolved in ethanol. The solution was left standing at room temperature for one day. The supernatant from the formed precipitate was taken off, concentrated under reduced pressure and the residue was lyophilized from water/acetonitrile to afford 15b with an enantiomeric excess of 99.6 % (e.e. determined by Chiral HPLC Chiralpak AS, mobile phase 0.2% TFA n-Hex/IPA) with spectroscopical properties identical to the compound described in Example 4, Step 1.
    [α]20 D = - 36.5±2.5°, (C = 0.50 in ethanol).
  • EXAMPLE 7 (-)-9-[[(Dimethylamino)sulfonyl](methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide
  • Figure imgb0019
  • Step 1 : (-)-9-[[(Dimethylamino)sulfonyl](methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide
  • To a solution of the amine synthesized in Example 5, Step 2 in methanol were added 4-fluoro-benzylamine (3 eq.) and triethylamine (2 eq.). The mixture was heated to 65 °C and stirred overnight. The solvent was removed under reduced pressure and the residue dissolved in dichloromethane. N,N-dimethylsulfamoyl chloride (3 eq.) and triethylamine (3 eq.) were added and the mixture was stirred for 4 h. The mixture was partitioned between ethylacetate and 0.1 M HCl. The aqueous phase was extracted with ethylacetate and the combined organic phases were dried over sodium sulfate, filtered and concentrated to dryness under reduced pressure. The product was purified by preparative RP-HPLC. using water (0.1 % TFA) and acetonitrile (0.1% TFA) as eluents (column: C18). The pooled product fractions were lyophilized to afford the title product as a fluffy white material (ee 90.7 %). The compound was dissolved in ethanol and left standing at room temperature for three days. The supernatant was taken off and concentrated to dryness under reduced pressure. The residue was redissolved in ethanol and the solution was left standing at room temperature for one day. The supernatant was taken off, concentrated under reduced pressure and the residue lyophilized from water/acetonitrile to afford the title product with an enantiomeric excess of 99.4 % (e.e. determined by Chiral HPLC Chiralpak AD, mobile phase 0.2% TFA n-Hex/IPA) with spectroscopical properties identical to the compound synthesized in Example 3, Step 4 except for the optical rotation.
    [α]20 D =33±1°, (C = 0.56 in ethanol).
  • EXAMPLE 8 (+)-9-[[(Dimethylamino)sulfonyl](methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide
  • Figure imgb0020
  • Step 1: (+)-9-[[(Dimethylamino)sulfonyl](methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide (16b)
  • The title compound was synthesized using as starting material the amine prepared in Example 6, Step 2 and following the procedure describe in Example 7, Step 1. The compound was obtained directly with the enantiomeric excess reported below and with spectroscopical properties identical to the compound synthesized in Example 3, Step 4 except for the optical rotation. Enantiomeric excess was determined by Chiral HPLC Chiralpak AD, mobile phase 0.2% TFA n-Hex/IPA.
    [α]20 D =+ 33±1°, (C = 0.69 in ethanol, ee 96.6 %).
  • EXAMPLE 9 (Reference) N-(4-Fluorobenzyl)-3-hydroxy-4-oxo-4,6,7,8-tetrahydropyrrolo[1,2-a]pyrimidine-2-carboxamide.
  • Figure imgb0021
  • Step 1: tert-butyl benzyloxy(3-cyanopropyl)carbamate (Bergeron, R. J., McManis, J. S., Tetrahedron 45(16), 4939-4944 (1989)).
  • The title comound was prepared according to the literature as described in EXAMPLE 1 -Step 1a, from 3-chloropropyonitrile.
    1H-NMR (CDCl3, 400 MHz, 300K) δ: 7.39-7-30 (m, 5H), 4.82 (s, 2H), 3.50 (t, J= 6.5 Hz, 2H), 2.31 (t, J = 7.2 Hz, 2H), 1.90-1.82 (m, 2H), 1.50 (s, 9H).
    13C NMR (CDCl3, 75 MHz, 300 K) δ 156.3,135.3, 129.5, 128.7,128.5, 119.1, 82.0, 77.0, 48.1, 28.3, 23.5, 14.9.
  • Step 2: 1-(benzyloxy)pyrrolidin-2-imine hydrochloride
  • The title compound was prepared from tert-butyl benzyloxy(3-cyanopropyl)carbamate as described in EXAMPLE 1- Step 2a.
    1H-NMR (DMSO-d6, 400 MHz, 300K) δ: 9.76 (s, 1H), 9.23 (s, 1H), 7.59-7.53 (m, 2H), 7.45-7.40 (m, 3H), 5.07 (s, 2H), 3.77 (t, J=7.1 Hz, 2H), 2.78 (t, J=7.7 Hz, 2H), 2.06-1.97 (m, 2H). 13C NMR (DMSO-d6, 75 MHz, 300K) δ 163.8, 133.7, 129.9, 129.3, 128.5, 75.9, 49.9, 26.8, 16.2.
    MS: m/z(%): 191 ((M+H)+, 100).
  • Step 3: 2-iminopyrrolidin-1-ol hydrochloride
  • The title compound was obtained from 1-(benzyloxy)pyrrolidin-2-imine hydrochloride as described for EXAMPLE 1- Step 3a.
    1H-NMR (DMSO-d6, 300 MHz, 300K) δ: 11.89 (s, 1H), 9.14 (s, 1H), 8.64 (s, 1H), 3.77 (t, J=7.3 Hz, 2H), 2.79 (t, J=7.9 Hz, 2H), 2.11-1.98 (m, 2H).
    13C NMR (DMSO-d6, 75 MHz. 300 K) δ161.3, 52.9, 26.8,16.1.
    MS: m/z(%): 101 ((M+H)+, 100), 83 (- H2O, 60).
  • Step 4: Methyl 3-hydroxy-4-oxo-4,6,7,8-tetrahydropyrrolo[1,2-a]pyrimidino-2-carboxylate.
  • To a solution of 2-iminopyrrolidin-1-ol hydrochloride in chloroform, triethylamine (1.5 eq) was added. The mixture was stirred for 5 min. at room temperature, then cooled to -30°C before the addition, dropwise under stirring, of dimethylacetylenedicarboxylate (1.1 eq) in chloroform. After 1h the solvent was removed under reduced pressure. To the resulting crude, 2 mL of anhydrous o-xylene was added and the mixture was heated under vigorous stirring at 150°C (oil bath temperature) for 2h, then the solvent was evaporated under reduced pressure. The residue was treated with MeOH and filtered, the filtrate was evaporated. The analytical sample was obtained by purification by preparative HPLC (Symmetry Column C18,5µm 19x300mm, gradient of CH3CN/H2O + 0.01% TFA).
    1H-NMR (DMSO-d6, 400 MHz, 300K) δ: 10.19 (s, 1H), 3.99 (t, J = 7.3 Hz, 2H), 3.80 (s, 3H), 2.92 (t, J = 7.9 Hz, 2H), 2.20-2.10 (m, 2H).
    13C NMR (DMSO-d6, 75 MHz, 300K) δ 166.4, 157.1, 153.6, 144.6,128.3, 52.1, 46.8, 30.9, 19.5.
    MS: m/z(%): 211 ((M+H)+, 100), 201 (100).
  • Step 5: Methyl 3-(benzoyloxy)-4-oxo-6,7,8,9-tetrahydropyrrolo[1,2-a]pyrimidine-2-carboxylate (A) and 4-oxo-4,6,7,8-tetahydropyrrolo[1,2-a]pyrimidin-3-yl benzoate (B)
  • To a solution of crude methyl 3-hydroxy-4-oxo-4,6,7,8-tetrahydropyrrolo[1,2-a]pyrimidine-2-carboxylate in pyridine benzoic anhydride (1.2 eq) was added and the mixture was stirred at room temperature overnight. The solvent was removed under reduced pressure and the residue was partitioned between ethyl acetate and NaHCO3 saturated solution. The organic phase is then washed with 1N HCl, brine, dried over Na2SO4, filtered and concentrated to dryness under vacuum. After flash chromatography of the oily residue on silica gel (eluent petroleum ether/EtOAc 1:3, then EtOAc) a solid was obtained, that by 1H NMR and mass proved to be a 2:1 mixture of product A and product B. Crystallization from EtOAc enriched the mixture to 6:1 A:B. Analytical samples of the two were obtained by prep.HPLC of the mixture (Symmetry Column C18, 5µm, 19x300mm, gradient of CH3CN/H2O + 0.01% TFA) and subsequent crystallization from EtOAc.
    A: 1H-NMR (DMSO-d6, 400 MHz, 300K) δ: 8.70 (d, J= 7.2 Hz, 2H), 8.29 (t, J= 7.4 Hz, 1H), 8.18-8.10 (m, 2H), 4.64 (t, J = 7.4 Hz. 2H), 4.31 (s, 3H), 3.64 (t, J = 8.1 Hz, 2H), 2.87-2.78 (m, 2H).
    13C NMR (DMSO-d6, 75 MHz, 300K) δ 163.1,163.1, 162.8, 156.0,142.4,136.0, 134.4,129.8, 129.1,127.8, 52.7, 47.6, 31.9,19.2.
    MS: m/z(%): 315 ((M+H)+, 100), 201 (25).
    m.p. 170.3-171.3 °C (EtOAc).
    B: 1H-NMR (DMSO-d6, 400 MHz, 300K) δ: 8.69 (d, J = 7.4 Hz, 2H), 8.42 (s, 1H), 8.27 (t, J= 7.4 Hz, 1H), 8.16-8.09 (m, 2H), 4.63 (t, J = 7.4 Hz, 210, 3.61 (t, J = 8.1 Hz, 2H), 2.86-2.76 (m, 2H).
    13C NMR (DMSO-d6, 75 MHz, 300K) δ 163.5, 163.3,155.5, 144.5,1363,134.2, 129.8, 129.0, 128.0, 47.2, 31.8, 19.4.
  • Step 6: N-(4-fluorobenzyl)-3-hydroxy-4-oxo-4,6,7,8-tetrahydrapyrrolo[1,2-a]pyrimidine-2-carboxamide.
  • To a solution of crude mixture of Methyl 3-(benzoyloxy)-4-oxo-6,7,8,9-tetrahydropyrrolo[1,2-a]pyrimidine-2-carboxylate and 4-oxo-4,6,7,8-tetrahydropyrrolo[1,2-a]pyrimidin-3-yl benzoate (ratio 6:1) in dry methanol, 3 eq. of 4-fluoro-benzylamine were added. The mixture was irradiated in a microwave apparatus (140 °C, 500 sec). After cooling the solvent was removed under reduced pressure. The product was isolated by preparative RP-HPLC (Symmetry Column C18, 5µm, 19x300mm, gradient of CH3CN/H2O + 0.01% TFA). The pooled product fractions were lyophilized to afford the title compound.
    1H-NMR (DMSOd6, 400 MHz, 300K) δ: 12.74 (bs, 1H), 8.96 (bs, 1H), 7.94-7.89 (m, 2H), 7.66-7.60 (m, 2H), 5.06 (d, J = 6.6 Hz, 2H), 4.56 (t, J = 7.2 Hz, 2H), 3.47 (t, J = 7.9 Hz, 2H), 2.77 (m, partially hidden under H2O).
    13C NMR (DMSO-d6, 75 MHz, 300 K) δ 168.6, 161.2 (d, J= 242 Hz), 156.3, 153.7, 147.0, 134.8 (d, J= 3 Hz), 129.5 (d, J= 8 Hz), 126.6; 115.0 (d, J= 21 Hz), 46.8, 41.4, 31.1, 19.3.
    MS: m/z (%): 304 ((M+H)+,100), 201 (70).
  • EXAMPLE 10 (Reference) 8-[[(Dimethylamino)sulfonyl](methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-4,6,7,8-tetrahydropyrrlo[1,2-a]pyrimidine-2-carboxamide.
  • Figure imgb0022
  • Crude methyl 3-(benzoyloxy)-4-oxo-4,6,7,8-tetrahydropyrrolo[1,2-a]pyrimidine-carboxylate, obtained as in EXAMPLE 9 - Step 5, was reacted as described in EXAMPLE 2 - Step 2. After purification by flash chromatography (eluent petroleum ether/ethyl acetate, 65:35) the resulting bromide (MS m/z: 3931395 (M+H)+) was obtained. The bromide was dissolved in anhydrous dimethylformamide and N-benzyl-N-methylamine (3 eq.) was added. The mixture was stirred for 2 h at room temperature and then concentrated to dryness under reduced pressure, to afford crude methylbenzylamine derivative (MS m/z: 330 (M+H)+). After dissolution in methanol, containing palladium on charcoal (10 % w/w), the compound was stirred under hydrogen at atmospheric pressure for 1.5 h. The catalyst was filtered and the solution was concentrated to dryness under reduced pressure to give crude methylamine derivative (MS m/z: 240 (M+H)+), which was dissolved in dry methanol. Triethylamine (2 eq) and 4-fluoro-benzylamine (3 eq) were added. The mixture was stirred and heated to 65 °C overnight. After cooling to room temperature the solvent was removed under reduced pressure. Crude p-fluorobenzylamide (MS m/z: 333 (M+H)+) was dissolved in dry dichloromethane and N,N-dimethylsulfamoyl chloride (3.5 eq.) was added. The mixture was stirred at room temperature for 16 h. The reaction was diluted with DCM and washed with HCl 1N and brine. The organic phase was dried on Na2SO4 and concentrated. The product was isolated by preparative RP-HPLC (Symmetry Column C18, 5µm, 19x300mm, gradient of CH3CN/H2O + 0.01% TFA). The pooled product fractions were lyophilized to afford the compound as a fluffy, slightly pink material.
    1H-NMR (DMSOd6, 400 MHz, 300K) δ: 12.42 (bs, 1H), 8.78 (t, J= 6.3 Hz, 1H), 7.42-7.35 (m, 2H), 7.23-7.15 (m, 2H), 5.27 (t, J= 9.1 Hz, 1H), 4.51 (ddd, J1= 6.3 Hz, J2= 14.8 Hz J3= 26.8 Hz, 2H), 4.13-4.05 (m, 1H), 3.88-3.72 (m, 1H, partially hidden under water), 2.78 (s, 6H), 2.73 (s, 3H), 2.48-2.38 (m, 1H), 2.35-2.23 (m, 1H).
    MS m/z: 440 (M+H)+.
  • EXAMPLE 11 (+/-)-N-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide
  • Figure imgb0023
  • The title compound was prepared according to the synthetic sequence described in EXAMPLE 4, with the following variations:
  • Step 1: 1-(Benzyloxy)azepan-2-imine
  • Tert-butyl-(benzyloxy)-(5-cyanopentyl)-carbamate (synthesized following the procedure described in EXAMPLE 1- Step 1a starting from 6-bromohexanenitrile) was dissolved in a saturated solution of HCl in ETCH and the mixture was stirred for 45 minutes. Nitrogen was bubbled into the solution to remove HCl in excess. The solvent was removed under reduced pressure and the residue, dissolved in 1,4-dioxane, was treated with triethylamine to adjust pH at 10. Ethanol was removed and the title compound containing an excess of triethylammonium chloride and ethyl 6-[(benzyloxy)amino]hexanoate was used without further purification. The analytical sample was purified by preparative RP-HPLC, using water (0.1% TFA) and acetonitrile (0.1% TPA) as eluents (column C18). The pooled product fractions were lyophilized.
    1H-NMR (400 MHz, DMSO-d6, 300K) δ: 9.39 (s, 1H), 8.81 (s,1H), 7.61-7.52 (m, 2H), 7.48-7.38 (m, 3H), 5.03 (s, 2H), 4.02-3.93 (m, 2H), 2.70-2.59 (m, 2H), 1.69-1.54 (m, 6H).
    MS m/z: 219 (M+H)+.
  • Step 2: (+/-)-N-(2-{[(4-Fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4-6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide
  • To a stirred solution of crude N-(4-fluorobenzyl)-3-hydroxy-10-(methylamino)-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxamide (synthesized starting from 1-(benzyloxy)azepan-2-imine according to the procedure used in the analogous 6-membered series (EXAMPLE 3 - Step 3) in dichloromethane were added 3 eq. of triethylamine, 2 eq of potassium (dimethylamino)(oxo)acetate, 2.2 eq. of 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride and 2.2 eq. of 1-hydroxybenzotriazole. The mixture was stirred at room temperature overnight The solvent was removed under reduced pressure and the residue was partitioned between ethyl acetate and 1M aq. HCl. The aqueous phase was extracted with ethyl acetate and the combined organic phases were dried over sodium sulfate, filtered and concentrated to dryness under vacuum. The title product was isolated by preparative RP-HPLC, using water (0.1 % TPA) and acetonitrile (0.1 % TPA) as eluents (column: C18). The pooled product fractions were lyophilized to afford the title compound as a fluffy, white material. The product is a mixture of rotamers by 1H NMR.
    1H-NMR (400 MHz, DMSO-d6, 300K) δ: 12.29 (bs, 0.1H), 11.95 (bs, 0.9H), 9.30 (bs, 0.9H), 8.45 (bt, 0.1H), 7.38 (dd, J = 8. 33, 5.5 Hz,1.8H), 7.33 (dd, J = 8.33, 5.5 Hz, 0.2H). 7.15 (t, J = 9.0 Hz, 2H), 5.45-5.25 (m, 0.9H), 4:94 (dd, J = 14, 5.7 Hz, 1.0H), 4.84-4.79 (m, 0.1H), 4.57-4.43 (m, 2H), 3.54 (dd, J = 14, 11 Hz, 0.9H), 3.28-3.18 (m, 0.1H), 3.05 (s, 0.3H), 2.92 (s, 2.7H), 2.90 (s. 5.4H), 2.81 (s, 0.3H), 2.76 (s, 0.3H), 2.19-1.78 (m, 5H), 1.41-1.27 (m, 1H).
    MS m/z: 460 (M+H)+.
  • EXAMPLE 12 (-)-N-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-ydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide Step.1: Dimethyl (2E)-2-[(azepan-2-ylidencamino)oxy]but-2-enedioate and dimethyl (2Z)-2-[(azepan-2-ylideneamino)oxy]but-2-enedioate
  • To a suspension of azepan-2-one oxime in acetonitrile 1.1 eq. of dimethyl acetylenedicarboxylate were added dropwise under stirring. The mixture was stirred at room temperature for 1 hour. The solvent was removed under reduced pressure to afford a mixture 8/1 of dimethyl (2E)-2-[(azepan-2-ylideneamino)oxy]but-2-enedioate and dimethyl (2Z)-2-[(azepan-2-ylideneamino)oxy]but-2-enedioate as a yellow oil. To better characterize the title compounds a small amount of the crude was purified by preparative RP-HPLC, using water (0.1% TPA) and acetonitrile (0.1% TFA) as eluents (column C18). The pooled product fractions were lyophilized Isomer E: 1H-NMR (400 MHz, DMSO-d6, 300K) δ: 7.08 (bs, 1H), 5.63 (s, 1H), 3.77 (s, 3H), 3.59 (s, 3H), 3.19-3.11 (m, 210, 2.29-2.21 (m, 2H), 1.66-1.42 (m, 6H).
    13C-NMR (125 MHz, DMSO-d6, 300 K) δ: 166.20, 162.81, 161.90, 161.61, 92.38, 52.42, 50.92, 42.28, 29.84, 29.32, 28.81, 25.40.
    MS m/z: 271 (M+H)+.
  • Isomer Z: 1H-NMR (400 MHz, DMSO-d6, 300K): 6.66 (bs, 1H), 5.63 (s,1H), 3.74 (s, 3H), 3.61 (s, 3H), 3.24-3.16 (m, 2H), 2.20-2.12 (m, 2H),1.65-1.44 (m, 6H).
    13C-NMR (125 MHz, DMSO-d6, 300 K) δ: 165.01, 163.01, 161.45, 154.11,101.09, 52.32, 51.05, 42.24, 29.93, 29.31, 28.45, 25.14.
    MS m/z :271 (M+H)+.
  • Step 2: Methyl 3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxylate
  • A mixture of dimethyl (2E)-2-[(azepan-2-ylideneamino)oxy]but-2-enedioate and dimethyl (2Z)-2-[(azepan-2-ylideneamino)oxy]but-2-enedioate in ratio 8/1 was dissolved in o-xylene and refluxed. After 16 h the solvent was removed under reduced pressure and the residue, dissolved in ethyl acetate, was extracted with a saturated solution of NaHCO3 in water. The pH of the aqueous phase was adjusted to about 3 adding 6M HCl aq. and the solution was extracted with dichloromethane. The organic phase was dried over sodium sulphate and concentrated. 1H-NMR (400 MHz, DMSO-d6, 300 K) δ: 10.12 (s, 1H), 4.29-4.16 (m, 2H), 3.80 (s, 3H), 2.95-2.78 (m, 2H), 1.79-1.41 (m, 6H).
    13C-NMR (100 MHz, DMSO-d6, 300K) δ:165.79, 158.54, 153.41, 143.55, 126.61, 5204, 43.02, 35.75, 28.76, 26.94, 24.58.
    MS m/z: 239 (M+H)+.
  • Step 3: Methyl 3-(benzoyloxy)-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxylate
  • To a solution of methyl 3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxylate in pyridine, benzoic anhydride (1.1 eq.) and a catalytic amount of dimethylaminopyridine were added. The mixture was stirred at room temperature for 3 h. The solvent was removed under reduced pressure and the residue partitioned between dichloromethane and 1M aq. HCl. The aqueous phase was extracted with dichloromethane and the combined organic phases were washed with 1M aq. HCl and brine. The organic phase was dried over sodium sulfate, filtered and concentrated to dryness under vacuum. Title compound was obtained after flash chromatography (eluents petroleum ether/ethyl acetate, 6:4) as a brown solid.
    1H NMR (400 MHz, DMSO-d6, 300 K) δ: 8.07 (dd, J = 8.6, 1.3 Hz, 2H), 7.78 (t, J = 7.5 Hz, 1H), 7.62 (t, J = 7.9 Hz, 2H), 4.31-4.29 (m, 2H), 3.74 (s, 3H), 3.06-3.04 (m, 2H), 1.82-1.65 (m, 6H). 13C-NMR (75 MHz, DMSO-d6, 300K) δ: 162.78, 162.65, 162.43,157.01, 140.29, 135.23, 134.18, 129.63, 128.86, 127.59, 52.46, 43.13, 36.07, 28.47, 26.12, 23.68.
    MS m/z: 343 (M+H)+.
  • Step 4: Methyl 3-(benzoyloxy)-10-bromo-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxylate
  • A mixture of methyl 3-(benzoyloxy)-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxylate, N-bromo-succininnide (2 eq.) and α,α'-azoisobutyronitrile ( 0.45 eq.) in carbon tetrachloride was stirred under reflux for 14 hour. The mixture was cooled to room temperature, the succinimide was filtered off and the solvent was removed under reduced pressure. Methyl 3-(benzoyloxy)-10-bromo-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxylate was obtained after flash chromatography (eluents petroleum ether/ethyl acetate, 8:2) as a pale yellow solid.
    1H-NMR (400 MHz, DMSO-d6, 300 K) δ: 8.07 (dd, J = 8.3, 0.9 Hz, 2H), 7.79 (t, J = 7.5 Hz, 1H), 7.63 (t, J = 7.9 Hz, 2H), 5.63 (dd, J = 5.9, 2.2 Hz, 1H), 4.98 (dd, J =14.3, 6.1 Hz, 1H), 3.97 (dd, J =14.3, 11.0 Hz, 1H), 3.76 (s, 3H), 2.31-2.13 (m, 2H), 2.10-1.79 (m, 3H), 1.-1.61-1.48 (m, 1H). 13C-NMR (75 MHz, DMSO-d6, 300 K) δ: 162.65, 162.14, 157.22, 157.10,139.45,136.59, 134.37,129.72,128.94,127.36, 53.56, 52.67, 42.37, 31.52, 25.78, 24.40.
    MS m/z: 423/421 (M+H)+.
  • Step 5: Methyl 3-hydroxy-4-oxo-10-{[(1R)-1-phenylethyl]amino}-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxylate
  • Methyl 3-(benzoyloxy)-10-bromo-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxylate (1.0 eq.) was added to a solution of (1R)-1-phenylethylamine (2.2 eq.) and triethylamine (1 eq) dissolved in N,N-dimethylformamide. The mixture was stained vigorously for 2 hours at room temperature and then at 50° C for 30 minutes. The solvent was removed under reduced pressure and the title crude product (a mixture 1:1 of diasteroiesomers) for use without further purification. In an alternative procedure (Step 5A), solid methyl 3-(benzoyloxy)-10-bromo-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxylate (1.0 eq.) was added to a solution of (1R)-1-phenylethylamine (45 eq.) dissolved in a 7:3 methanol/water mixture at -30°C. The reaction was carried out over night, then the temperature was raised to room temperature and the solvent concentrated to obtain a white solid that was filtered off and discarded. The title compound ( as a mixture 7:3 of diasteroisomers) was extracted in dichloromethane from the mother liquid for use in the next step without further purification.
    MS m/z: 358 (M+H)+.
  • Step 6: N-(4-fluorobenzyl)-3-hydroxy-4-oxo-10-{[(1R)-1-phenylethyl]amino}-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxamide
  • p-Fluorobenzylamine (3 eq.) was added to the methyl 3-hydroxy-4-oxo-10-{[(1R)-1-phenylethyl]amino}-4,b,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxylate (prepared as described in Step 5 or Step 5A) dissolved in methanol. The mixture was stirred at 70° C overnight, then cooled to room temperature for use directly in Step 7. In an alternative procedure (Step 6A), the solvent was removed and the crude product crystallized from acetonitrile several times to obtain a single diasteroisomer of the title compound as the 4-fluombenzylam=nonium salt with d.e.>95%
  • Step 7: (+) N-(4-fluorobenzyl)-3-hydroxy-10-{methyl[(1R)-1-phenylethyl]amino}-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxamide
  • N-(4-Fluorobenzyl)-3-hydroxy4-oxo-10-([(1R)-1-phenylethyl]amino)-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxamide obtained in Step 6 was dissolved in methanol and the pH was adjusted to circa 5 with acetic acid before 37 % aqueous formaldehyde (6 eq.) and sodium cyanoborohydride (6.25 eq.) were added. The mixture was stirred at room temperature overnight. The solvent was removed under reduced pressure and the residue, dissolve in the minimum amount of methanol, was acidified to pH 2-3 with trifluoroacetic acid The solution was applied on cation-exchange resin cartridges (Varian MEGA BOND ELUTE SCX), the cartridges washed with methanol and the crude product was eluted with 2 M ammonia in methanol. The pooled eluents were concentrated to dryness under reduced pressure to get an oily residue. The product, a mixture of diasteroisomers, was separated by preparative RP-HPLC-purification (column: C18) eluants water (0.1 % TFA), acetonitrile (0.1 % TFA). The title diasteroisomer was eluted as the first peak, and after lyophilization the title compound was obtained as a white solid (TFA salt).
  • In an alternative procedure (Step 7A), the product of Step 6A was reacted in the same manner as the product of Step 6 to obtain a single diastereomer without separation by HPLC.
    1H-NMR (300 MHz, DMSO-d6-TFA, 300 K) 8: 9.42 (t, J = 6.2 Hz, 1H), 9.20 (bs, 1H), 7.60 (bd, J = 7.3 Hz, 2H), 7.51-7.29 (m, 5H, 7.21 (t, J = 8.9 Hz, 2H), 4.98-4.75 (m, 3H), 4.69 (dd, J = 15.5, 6.9 Hz, 1H), 4.47 (dd, J = 15.5, 5.5 Hz, 1H), 3.66 (t, J =12.8 Hz, 1H), 2.94-2.81 (m, 3H), 1.97-1.81 (bm, 2H), 1.79-1.33 (m, 7H).
    MS m/z: 465 (M+H)+.
    [α]20 D = + 62 ± 2 (C = 0.1 in chloroform).
  • Step 8: (-)2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-N-methyl-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-ammonium trifluoroacetate
  • A solution of the TFA salt of the product of Step 7 (or 7A) in methanol, containing palladium on charcoal (10%, w/w) was stirred under hydrogen at atmospheric pressure for 4 h. The catalyst was filtered off and the solution was concentrated to dryness under reduced pressure to afford the title compound.
    1H-NMR (300 MHz, DMSO-d6- TPA, 300K) 8: 9.88 (bs, 1H), 9.56 (bs, 1H), 9.14 (bs, 1H), 7.41 (dd, J = 8.6, 5.7 Hz, 2H), 7.17 (t, J = 8.8 Hz, 2H), 4.92 (dd, J =14.6, 4.6 Hz, 1H), 4.72 (bm, 1H), 4.58-4.44 (m, 2H), 3.51 (dd, J =13.9, 11.7 Hz, 1H), 2.66 (t, J = 4.9 Hz, 3H), 2.29 (d, J = 13.3 Hz, 1H), 2.02-1.57 (m, 4H),1.45-1.27 (m, 1H).
    MS m/z: 361 (M+H)+.
    [α]20 D = - 4 ± 2 (C = 0.4 in methanol).
  • Step 9: (-)N-(-{[(4-Fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide methyl chlorooxoacetate (2-6 eq.) and N-ethyldiisopropylamine (4 eq.) were added to a solution of the ammonium tritfluoroacetate compound of Step 8 in chloroform. The mixture was stirred at room temperature for 1 hour. The solvent was removed under reduced pressure and the residue dissolved in a 2 M solution of dimethylamine in methanol was stirred at room temperature for 6 hours. The solvent was removed under reduced pressure and the residue dissolved in dichloromethane was washed with 1 M HCl in water. The organic phase was dried over anhydrous Na2SO4 and the solvent removed under reduced pressure. The title product was isolated by preparative RP-HPLC, using water (0.1% TFA) and acetonitrile (0.1% TFA) as eluents (column: C18). The pooled product fractions were lyophilized to afford the title product with an enantiomeric excess of 99.5% (e.e. determined by Chiral HPLC Chiralpak AD, mobile phase 0.2% TFA n-Hex/0.2% TFA ethanol with 3% methanol). An amorphous potassium salt of the title compound was obtained by treating the compound dissolved in acetonitrile with aqueous KOH and then freeze drying.
    1H-NMR spectra was identical to the compound described in Example 11.
    13C-NMR (100 MHz, DMSO-d6, 300 K) δ: 168.01, 165.80, 165.03, 161.30 (d, JC-F = 243 Hz), 157.68, 149.67, 145.94, 134.59, 12956 (d, JC-F = 8.5 W), 124.72, 115.10 (d, JC-F = 21 Hz), 55.88, 42.42, 41.56, 36.24, 32.79, 32.34, 28.83, 27.10, 26.15.
    MS m/z: 460 (M+H)+.
    [α]20 D = - 72 ± 2 (C = 0.1 in chloroform).
  • EXAMPLE 13 Racemic N-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide Step 1: Preparation of ω-Hydroxy N-Methyl aminonitrile 3
  • Figure imgb0024
    Materials MW Equiv. Moles Wt. (g) Vol. (mL) Density
    DHP 84.12 1 0.2500 21.10 22.93 0.92
    5% H2SO4 98.08 0.122 0.0305 60 mL
    40% MeNH2 31.06 0.244 0.0610 4.74 5.3 0.902
    MeNH2.HCl 67.51 5 1.250 84.4
    NaCN 49.01 1 0.2500 12.25
    IPAc 900
  • To a 5% H2SO4 aqueous solution was added 3,4-dihydro-2H-pyran (DHP; 21.1 g) at 20-35 °C. The resulting solution was aged at 20-35 °C for 1 h. The reaction mixture was cooled to 0-5 °C, and neutralized to pH = 6-7 by 40% aqueous methylamine. Methylamine hydrochloride and sodium cyanide were added respectively to the reaction mixture. The resulting solution was aged at room temperature for 36 h. The reaction mixture was extracted by IPAc (6 x 150 mL). The combined organic layers were concentrated to a total volume about 150 mL (assay yield about 91 %) and was used in the next step. 1H NMR (CDCl3, 400 MHz) δ: 3.81 (m, 1H), 3.45 (m, 2 H), 2.47 (s, 3 H), 1.90-1.40 (m, 6 H).
  • Step 2: Preparation of ω-Hydroxy N-Methyl N-Boc-aminonitrile 4
  • Figure imgb0025
    Materials MW Equiv. Moles Wt. (g) Vol. (mL)
    Aminonitrile 3 142.20 1 0.2106 29.95
    (Boc)2° 218.25 1.05 0.2211 48.3
    5% NH2OH/ 35
    10%NH4Cl
    IPAc 80
  • To a solution of ω-hydroxy N-methyl aminonitrile 3 (0.2106 moles) in IPAc (from Step 1) was added (Boc)2O (48.3 g) at room temperature. The resulting solution was aged at 30-35 °C for 2 h (100% conversion by 1H NMR). The reaction mixture was cooled to 0-5 °C and 5%NH2OH/10%NH4Cl (35 mL) was added. The resulting mixture was aged at 10-20 °C for 3 h. After a phase cut, the aqueous layer was extracted with IPAc (80 mL), the combined organic layers were washed with brine (50 mL), and then concentrated and solvent-switched to IPA (total volume 230 mL), which was used for next step. 1H NMR (CDCl3, 400 MHz) δ: 5.18 (m, 1 H), 3.64 (q, J = 5.7 Hz, 2 10, 2.88 (s, 3 H). 1.88-1.75 (m, 3 H). 1.65-1.61 (m, 2 H), 1.49-1.46 (m, 1 H), 1.18 (s, 9 H).
  • Step 3: Preparation of Hydroxyamidine 5
  • Figure imgb0026
    Materials MW Equiv. Moles Wt. (g) Vol. (mL) Density
    N-Boc-aminonitrile 4 242.31 1 0.2106 51.03
    50% NH2OH 33.03 1.25 0.2633 17.40 16.20 1.078
    IPA 180
    MeOH 600
  • To a solution of N-Boc-aminonitdle 4 (0.2106 moles) in IPA. (total volume 230 mL) was added 50% hydroxylamine (16.2 mL) at ambient temperature. The resulting solution was aged at 60 °C for 3 h. The reaction mixture was then concentrated and solvent-switched to methanol solution (total volume 230 mL), which was used in the next step. 1H NMR (CDCl3, 400 MHz) δ: 7.53 (br s, 1H), 4.84 (br s, 2 H), 4.64 (t. J = 7.1 Hz, 1 H), 3.71-3.62 (m, 2 H), 2.72 (s, 3 H), 2.00 (br s, 1 H), 1.92-1.82 (m, 1H), 1.76 (1.55 (m, 3 H), 1.49 (s, 9 H), 1.42-1.23 (m, 2 H).
    HPLC conditions: Column: Zorbax, Rx C8 250 x 4.6 mm; Temperature: 30 ° C; Detection at 210 nm; Mobile Phase: 0.1% aq H3PO4 (A)/MeCN (B); Gradient: 90:10 (A)/(B) to 10:90 over 15 min, 10:90 hold for 5 min,10:90 to 90:10 (A)/(B) over 10 seconds; Flow Rate: 1 mL/min. Retention time: amidoxime - 6.152 minutes and 6.256 minutes (two isomers)
  • Step 4: Preparation of 0-Alkene Amidoxime 6
  • Figure imgb0027
    Materials MW Equiv. Moles Wt. (g) Vol. (mL) Density
    Hydroxyamidine 5 275.35 1 0.2106 57.93
    DMAD 142.11 1.05 0.2211 31.42 27.10 1.16
    MeOH
    Cumene 500
  • To a solution of hydroxyamidine 5 (about 0.2106 mole) in methanol (total volume 230 mL) was added dimethyl acetylenedicarboxylate (27.10 mL) at room temperature. The resulting solution was aged at room temperature for 16 h. The reaction mixture was concentrated and solvent-switched to cumene at 40-60 °C (total volume 430 mL). The solution was used in the next step.1H NMR (CDCl3, 400 MHz) δ: 5.82 (s 0.28 H), 5.73 (s, 0.72 H), 5.44 (br s, 1.77 H), 5.25 (br s, 0.56 H), 4.61 (m, 1 H), 3.89 (s, 0.84 H), 3.84 (s, 2.16 H), 3.72 (s, 2.16 H), 3.68 (s, 0.84 H), 3.65-3.58 (m, 2H), 2.73 (s, 0.84 H), 2.71 (s, 2.16 H), 1.90-1.52 (m, 4 H), 1.47 (s, 9 H), 1.43-1.30 (m, 2 H).
    HPLC conditions: Column: Zorbax, Rx C8 250 x 4.6 mm; Temperature: 30 ° C; Detection at 210 nm; Mobile Phase: 0.1% aq H3PO4 (A)/eoCN (B); Gradient: 90:10 (A)/(B) to 10:90 over 15 min, 10:90 hold for 5 min. 10:90 to 90:10 (A)/(B) over 10 seconds; Flow Rate: 1 mL/min. Retention time: amidoxime 6 - 12.051 minutes, 12.315 minutes, ratio ca 3.6: 1.
  • Step 5: Preparation of Pyrimidine 7
  • Figure imgb0028
    Materials MW Equiv. Moles Wt. (g) Vol. (mL) Density
    O-Alkene 417.45 1 0.2106 87.91
    amidoxime 6
    Cumene 430 (total)
    5% NaHCO3 84.1 1.44 0.3032 510
    EtOAc 750
    Brine 150
    THF
  • A solution of O-alkene amidoxime 6 (about 0.2106 moles) in cumene (total volume 430 mL) was heated at 120 °C (inside temperature) for 12 h. The reaction mixture was then cooled to about 60 °C, concentrated to a total volume 250 mL, then diluted with EtOAc (250 mL), and cooled to 25-35 °C. 5% Sodium bicarbonate (330 mL, about 1 equiv.) was then slowly added, and the resulting solution was aged at 25-35 °C for 0.5 h. After a phase cut, the organic layer was extracted with 5% sodium bicarbonate (180 mL) again. The combined aqueous extracts were acidified by 5 N HCl to pH = 2-3, and extracted by EtOAc (3 x 250 mL). The combined organic layers were washed with brine (150 mL). The organic solution was concentrated and solvent-switched to THF (about 30-40% yield overall, KF about 100-150 ppm). 1H NMR (CDCl3, 400 MHz) δ: 10.66 (br s, 2 H), 4.77 (m, 1 H), 4.01 (s, 3H), 3.72-3.67 (m, 2 H), 2.77 (s, 3 H), 2.20-1.55 (m, 5 H), 1.48 (s, 9 H), 1.43-1.35 (m, 1 H).
    HPLC conditions: Column: Zorbax, Rx C8 250 x 4.6 mm; Temperature: 30 ° C; Detection at 210 nm; Mobile Phase: 0.1% aq H3PO4 (A)/MeCN (B); Gradient: 90:10 (A)/(B) to 10:90 over 15 min, 10:90 hold for 5 min, 10:90 to 90:10 (A)/(B) over 10 seconds; Flow Rate: 1 mL/min. Retention time: pyrimidine 7 - 9.905 minutes
  • Step 6: Preparation of Bismesyl-Pyrimidine 8
  • Figure imgb0029
    Materials MW Equiv. Moles Wt.(g) Vol. (mL) Density
    Pyrimidine 7 385.41 1 0.09029 43.5 (80%)
    TEA 101.19 3 0.2709 27.4 37.8 0.726
    MsCl 114.55 3 0.2709 31.0 21.0 1.480
    THF 575
    K2CO3 138.21 1 0.09029 12.5
    MeOH 200
    EtOAc 400
  • To a solution of pyrimidine 7 (43.5 g, about 80% pure, 0.09029 moles) in THF (275 m L) was slowly added TEA (37.8 mL) and MsCl (21.0 mL) at the same time at 0-5 °C over 1 h. The resulting solution was aged at the same temperature for 4 h. The solid was filtered off, washed with THF (3 x 100 mL). The combined filtrations were concentrated and solvent-switched to methanol (total volume 200 mL). To the trimesyl-pyrimidine in methanol solution was added potassium carbonate (12.5 g, 0.09029 moles) at 10-20 °C. The resulting solution was aged at the same temperature for 6-10 h (monitored by HPLC). The reaction mixture was neutralized to pH = 6-7 by 5 N HCl, and concentrated to a total volume about 100 mL. 16% brine (100 mL) was added, and the resulting solution was extracted by EtOAc (3 x 100 mL). The combined organic layers were washed with brine (50 mL), concentrated and solvent-switched to DMF. The by-product (MeSO3Me), which was generated in 1 equiv from the selectively hydrolysis of the trimesyl-pynmidine, was removed by azeotrope with DMF at 60-65 °C (monitored by 1H NMR until <10 mole%). The concentration of bismesyl-pyrimidine 8 in DMP was about 0.3 M (total volume 300 mL). 1H NMR (CDCl3, 400 MHz) δ: 11.00 (br s, 1 H), 4.78 (d, J= 7.8 Hz, 1 H), 4.24-4.15 (m, 2 H), 3.95 (s, 3 H), 3.50 (s, 3 H), 2.99 (s, 3H), 2.81 (s, 3 H), 2.12-2.11 (m, 1 H), 1.90-1.76 (m, 2 H), 1.46 (s, 9 H), 1.43-1.35 (m, 2 H).
    HPLC conditions: Column: Zorbax, Rx C8 250 x 4.6 mm; Temperature: 30 ° C; Detection at 210 nm; Mobile Phase: 0.1% aq H3PO4 (A)/MeCN (B); Gradient: 90: 10 (A)/(B) to 10:90 over 15 min, 10:90 hold for 5 min, 10:90 to 90:10 (A)/(B) over 10 seconds; Flow Rate: 1 mL/min. Retention time: trimesyl-pyrimidine - 14.140 minutes; bismesyl-pyrimidine 12.760 minutes.
  • Step 7: Preparation of Seven-Membered Ring Pyrimidine Mesylate 9
  • Figure imgb0030
    Materials MW Equiv. Moles Wt. (g) Vol. (mL)
    Bismesylpyrimidine 8 541.59 1 0.09029 48.90
    Cs2CO3 325.82 1.2 0.1083 35.30
    DMF
  • To a solution of bismesyl-pyrimidine 8 (0.09029 moles) in DMF (total volume 300 mL) was added cesium carbonate (35.30 g) at room temperature. The resulting slurry was aged at 55 °C for 2-3 h (76% conversion by HPLC). After being neutralized to pH = 7, the reaction mixture was diluted with 250 mL of water, extracted with IPAc (2 x 250 mL). The combined organic layers were washed with brine (2 x 200 mL). The organic layer was concentrated to give crude product. Half of the crude product was purified by passing a short column (silica gel, hexane: EtOAc 2: 1) to afford desired product 9 (6.00 g, 98A% pure), and 9a (2.3 g, 40A% pure). The overall yield from DHP to cyclized product is about 13% after collection.
    1H NMR (CDCl3, 400 MHz) For compound 9 : 8: 5.34 (m, 1 H), 5.22 (m, 1 H), 3.93 (s, 3 H), 3.51 (s, 3 H), 3.47 (m,1H), 2.97 (s, 3 H), 2.20-2.05 (m, 3 H), 1.90-1.65 (m, 2 H), 1.44 (s, 9 H), 1.24 (m, 1 H). For compound 9a : 11.86 (br s, 1 H), 7.90-7.55 (br s, 1H), 7.31 (dd, J = 8.5, 5.4 Hz, 2 H), 7.06 (t, J = 8.5 Hz, 2 H), 5.40-4.90 (m, 2 H), 4.53-4.40 (m, 2 H), 3.45-3.23 (m,1 1H). 2.23-2.05 (m, 3 H), 1.86.-1.76 (m, 1 H), 1.74-1.64 (m, 1 H), 1.47-1.37 (m, 1 H), 1.30 (s, 9 H). HPLC conditions: Column: Zorbax, Rx C8 250 x 4.6 mm; Temperature: 30 ° C; Detection at 210 nm; Mobile Phase: 0.1% aq H3PO4 (A)/MeCN (B); Gradient: 90: 10 (A)/(B) to 10:90 over 15 min, 10:90 hold for 5 min, 10:90 to 90:10 (A)/(B) over 10 seconds; Flow Rate: 1 mL/min. Retention time: the seven-membered ring-pyrimidine mesylate 9: 13.969 minutes; the seven-membered ring-pyrimidine 9a : 13.141 minutes.
  • Alternative procedure using LiH was also employed: To a solution of bismesyl-pyrimidine 8 (65 mg) in dioxane (1 mL) was added LiH powder at room temperature. The resulting mixture was aged at 65 ° C for 4 h. The reaction mixture was then cooled to room temperature and 1 N HCl was added to quenched the excess LiH. The solution was extracted with EtOAc (2 x 5 mL). The combined organic layer was washed with brine, and then concentrated. The residue was purified by flash chromatography (silica gel, hexane:EtOAc = 2:1) to afford seven-membered ring pyrimidine mesylate 9 (45.6 mg, 85%). 1H NMR (CDCl3, 400 MHz) 8: 5.34 (m, 1 H), 5.22 (m, 1 H), 3.93 (s, 3 H), 3.51 (s, 3 H), 3.47 (m, 1 H), 2.97 (s, 3 H), 2.20-2.05 (m, 3 H), 1.90-1.65 (m, 2 H), 1.44 (s, 9 H), 1.24 (m, 1 H).
  • Step 8: Preparation of Seven-Membered Ring-Pyrimidine Amide 10
  • Figure imgb0031
    Materials MW Equiv. Moles Wt (g) Vol. (mL) Density
    Seven-membered ring-pyrimidine 9 445.49 1 0.01347 6.000
    4-fluorobenzylamine 125.15 3 0.04041 5.060 5.22 1.09
    EtOH 80
  • To a solution of seven-membered ring-pyrimidine mesylate 9 (6 g) in EtOH (80 mL) was added 4-fluorobenzylamine (5.060 g). The resulting solution was reflux for 8 h. (100% conversion by HPLC). The reaction mixture was concentrated to about 20 mL total volume, and 80 mL of EtOAc was added. To the resulting solution was added 20% brine (15 mL), 4 N HCl (15 mL), and water 10 mL). After a phase cut, the aqueous layer was back-extracted with EtOAc (25 mL). The combined organic layers were washed with 4 N HCl : 20% brine (1:1,3 x 15 mL), brine (15 mL). The organic solution was concentrated to a total volume about 30 mL Hexane (70 mL) was slowly added to the solution over 1 h. The resulting slurry was aged at 0-5 °C for 1 h. The crystalline solid was filtered off, washed with hexane:EtOAc (4:1, 50 mL), dried under vacuum with nitrogen sweep to afford seven-membered ring-pyrimidine amide 10 (5.30 g, 86%, HPLC >97A%).1H NMR (CDCl3, 400 MHz) 8: 11.85 (br s, 1H), 7.84 (br s, 0.5 H), 7.68 (br s, 0.5 H), 7.31 (m, 2 H), 7.04 (m, 2 H), 5.40-4.90 (m, 2 H), 4.53 (m, 2 H), 3.38 (m, 1H), 2.87 (s, 3 H), 2.20-2.15 (m, 3 H), 1.90-1.40 (m, 3 H), 1.37 (s, 9 H).
    HPLC conditions: Column: Zorbax, Rx C8 250 x 4.6 mm; Temperature: 30 ° C; Detection at 210 nm; Mobile Phase: 0.1% aq H3PO4 (A)/MeCN (B); Gradient: 90:10 (A)/(B) to 10:90 over 15 min, 10:90 hold for 5 min, 10:90 to 90:10 (A)/(B) over 10 seconds; Flow Rate: 1 mL/min. Retention time: the seven-membered ring-pyrimidine 10 - 15.467 minutes.
  • Step 9: Preparation of Seven-Membered Ring-Pyrimidine Amide Hydrochloride Salt 11
  • Figure imgb0032
    Materials MW Equiv. Moles g mL Density
    Seven-membered 460.50 1 0.001846 0.8500
    pyrimidine amide 10
    HCl (gas) 36.46 8 0.01478 0.5389
    EtOAc 3.5
  • To a solution of ethyl acetate (3.5 mL) was bubbled HCl gas (0.5389 g), at -30 to -20 °C. N-Boc-seven-membered ring pyrimidine amide 10 (crystalline solid) was charged to the HCl-EtOAc solution at -30 to -20 °C. The resulting solution was slowly warmed to room temperature over 2.5 h, and aged at room temperature for 0.5 h (100% conversion by HPLC). The reaction mixture was diluted by EtOAc (7 mL). The resulting slurry was aged at 0-5 °C for 1 h. The crystalline solid was filtered off, washed with EtOAc, hexane, dried under vacuum with nitrogen sweep to afford desired product 11 (98% isolated yield, >97A % pure). 1H NMR (CDCl3, 400 MHz) δ: 12.35 (s, 1 H), 9.96 (t, J= 6.3 Hz, 1H), 9.51 (br s, 1 H), 9.19 (br s, 1 H), 7.42 (dd, J = 8.5, 5.6 hz, 2 H), 7.19 (t, J = 8.5 Hz, 2 H), 4.92 (dd, J =14.5, 5.1 Hz, 1 H), 4.71 (m, 1 H), 4.57-4.45 (m, 2 H), 3.52 (t, J = 14.5 Hz), 2.65 (t, J = 5.0 HZ, 3 H), 2.30 (br d, J = 12.6 Hz, 1 H), 1.99-1.92 (m, 1 H), 1.90-1.75 (m, 2H), 1.68-1.60 (m, 1H),1.41-1.33 (m, 1 H).
    HPLC conditions: Column: Zorbax, Rx C8 250 x 4.6 mm; Temperature: 30 ° C; Detection at 210 nm; Mobile Phase: 0.1% aq H3PO4 (A)/MeCN (B); Gradient: 90:10 (A)/(B) to 10:90 over 15 min, 10:90 hold for 5 min, 10:90 to 90:10 (A)/(B) over 10 seconds; Flow Rate: 1 mL/min. Retention time: the seven-membered ring-pyrimidine hydrochloride salt 11 - 8.118 minutes.
  • Steg 10: Preparation of Racemic N-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide 14
  • Figure imgb0033
    Materials MW Equiv. mmoles Wt. (g) Vol. (mL) Density
    Acid 12 (96% pure) 117.10 5 1.000 0.122
    Ethyl chloroformate 125.15 4.8 0.960 0.104 0.092 1.135
    4-NMM 101.15 4.8 0.960 0.0971 0.106 0.9200
    THF 3
    pyrimidine 396.84 1 0.200 0.0794
    hydrochloride salt 11
    40% dimethylamine 45.07 6.25 1.250 0.141 0.158 0.8900
    2 NHCl
  • To a solution of acid 12 (122 mg) in THF (3 mL) was added ethyl chloroformate (92 µl) at 0-5 °C. Then, 4-NMM (106 µl) was slowly added to the reaction mixture at 0-5 °C. The reaction mixture was aged at the same temperature for 2 h. The pyrimidine hydrochloride salt 11 (79.4 mg) was added as a solid to the mixed-anhydride solution at 0-5 °C, and aged at the same temperature for 5 h, and then at 5-10 °C for another 2 h (100% conversion by HPLC). Dimethylamine aqueous (40%, 158 µl) was added to the reaction mixture, and the mixture aged at 10-15 °C for 1 h, wherein the reaction was monitored by HLPC to assure complete conversion. The reaction mixture was acidified by 2 N HCl to adjust to pH = 3-4 at 5-15 °C. EtOAc (6 mL) and brine (2 mL) were added, respectively. After phase cut, the organic layer was washed with 1 N HCl (2 mL), brine (2 x 2 mL). The organic layer was concentrated to a total volume of 1 mL. Hexane (5 mL) was slowly added over 0.5 h. The resulting slurry was aged at 0-5 °C for 1 h. The crystalline solid was filtered off, washed with hexane/EtOAc (5: 1), MTBE, dried under vacuum with nitrogen sweep to give the title compound 14 (75.6 mg, 82%). 1H NMR (CDCl3, 400 MHz) δ: 12.13 (s, 1H), 9.41 (br s, 1 H), 7.38 (dd, J = 8.5, 5.4 Hz, 2 H), 7.00 (t, J = 8.5 Hz, 2 H), 5.40 (br s, 1H), 5.29 (dd, J = 14.5, 6.0 Hz, 1 H), 4.60 (dd, J =14.5, 6.6 Hz, 1H), 4.52 (dd, J = 14.5, 6.3 Hz, 1 H), 3.35 (dd, J = 14.5, 11.6 Hz, 1H), 3.04 (s, 3 H), 3.01 (s, 3 H), 2.98 (s, 3 H), 2.23-2.12 (m, 3 H), 1.95-1.81 (m, 2H), 1.58-1.49 (m, 1H).
    HPLC conditions: Column: Zorbax, Rx C8 250 x 4.6 mm; Temperature: 30 ° C; Detection at 210 nm; Mobile Phase: 0.1% aq H3PO4 (A)/MeCN (B); Gradient: 90:10 (A)/(B) to 10:90 over 15 min, 10:90 hold for 5 min, 10:90 to 90:10 (A)/(B) over 10 seconds; Flow Rate: 1 mL/min. Retention time: the title compound 14 - 12.191 minutes.
  • Table 1 below lists Compounds of the present invention which have been prepared. The table provides the structure and name of each compound, the mass of its molecular ion plus 1 (M+) or molecular ion minus 1 (M-) as determined via FIA-MS, and the synthetic scheme employed to prepare the compound.
    Structure Name M+ Scheme
    Figure imgb0034
    N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(methylsulfonyl)acetyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 467 C
    Figure imgb0035
    N-(4-fluorobenzyl)-3-hydroxy-9-morpholin-4-yl-4-oxo-6,7,8,9-tetrahydro-4H-pyrrido[1,2-a]pyrimidine-2-carboxamide 403 B
    (Ex. 2)
    Figure imgb0036
    N1-(2-{[(4-fluorobenzyl)amino]-carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N1,N2,N2-trimethylethanediamide 446 C
    (Ex. 4)
    Figure imgb0037
    9-[[(dimethylamino)sulfonyl]-(methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 454 C
    (Ex. 3)
    Figure imgb0038
    N-(4-fluorobenzyl)-3-hydroxy-9-{[(methylsulfonyl)acetyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 453 C
    Figure imgb0039
    (+)-9-[[(dimethylamino)sulfonyl]-(methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 454 D
    (Ex. 8)
    Figure imgb0040
    (-)-9-[[(dimethylamino)sulfonyl]-(methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 454 D
    (Ex. 7)
    Figure imgb0041
    (+)N1-(2-{[(4-fluorobenzyl)amino]-carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N1,N2,N2-trimethylethanediamide 446 D
    (Ex. 5)
    Figure imgb0042
    (-)N1-(2-{[(4-fluorobenzyl)amino]-carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N1,N2,N2-trimethylethanediamide 446 D
    (Ex. 6)
    Figure imgb0043
    (+)-N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(methylsulfonyl)acetyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 467 D
    Figure imgb0044
    (-)-N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(methylsulfonyl)acetyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 467 D
    Figure imgb0045
    N-(2-{[(4-fluoro-3-methylbenzyl)-amino]carbonyl]-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide 460 C
    Figure imgb0046
    N-(2-{[(3-chloro-4-methylbenzyl)-amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide 476 C
    Figure imgb0047
    N-(2-{[(3-chlorobenzyl)amino]-carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide 462 C
    Figure imgb0048
    N-(4-fluorobenzyl)-3-hydroxy-9-(6-mathyl-1,1-dioxido-1,2,6-thiadiazinan-2-yl)-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 466 B
    Figure imgb0049
    (-)-N-(2-{[(4-fluoro-3-methylbenzyl)-amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide 460 D
    Figure imgb0050
    (-)-N-(2-{[(3-chloro-4-methylbenzyl)-amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide 476 D
    Figure imgb0051
    (+)-N-(4-fluorobenzyl)-3-hydroxy-9-(6-methyl-1,1-dioxido-1,2,6-thiadiazinan-2-yl)-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 466 D
    Figure imgb0052
    N-(4-fluorobenzyl)-3-hydroxy-9-[methyl(pyrrolidin-1-ylsulfonyl)amino]-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 480 C
    Figure imgb0053
    9-[(azetidin-1-ylsulfonyl)(methyl)-amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 466 C
    Figure imgb0054
    (-)-N-(4-fluorobenzyl)-3-hydroxy-9-[methyl(morpholin-4-ylsulfonyl)-amino]-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 496 D
    Figure imgb0055
    (+)-N-(4-fluorobenzyl)-3-hydroxy-9-[methyl(morpholin4-ylsulfonyl)-amino]-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 496 D
    Figure imgb0056
    (+)9-[(azetidiD-1-ylsulfonyl)(methyl)-amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 466 D
    Figure imgb0057
    (+)-N-(4-fluoro-3-methylbenzyl)-3-hydroxy-9-[methyl(morpholin-4-ylsulfonyl)amino]-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 510 D
    Figure imgb0058
    (-)-N-(4-fluorobenzyl)-3-hydroxy-9-(methyl[(4-methylpiperazin-1-yl)sulfonyl]amino)-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 509 D
    Figure imgb0059
    (+)-N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(4-methylpiperazin-1-yl)sulfonyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 509 D
    Figure imgb0060
    N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(4-methylpiperazin-1-yl)sulfonyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide 509 D
    Figure imgb0061
    N-(2-{[(4-fluorobenzyl)amino]-carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide 460 C
    (Ex. 11)
    Figure imgb0062
    (-)N-(2-{[(4-fluorobenzyl)amino]-carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide 460 D
    (Ex. 12)
    Figure imgb0063
    (+)N-(2-{[(4-fluorobenzyl)amino]-carbonyl}-3-hydroxy-4-oxo-4,6,7,8,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide 460 D
    Figure imgb0064
    N-(2-{[(4-fluoro-3-methylbenzyl)-amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide 474 C
    Figure imgb0065
    N-(2-{[(3-chloro-4-methylbenzyl)-amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide 490 C
    Figure imgb0066
    (+)-N-(2-{[(4-fluoro-3-methylbenzyl)-amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl-N,N',N'-trimethylethanediamide 474 D
    Figure imgb0067
    (-)-N-(2-{[(4-fluoro-3-methylbenzyl)-amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide 474 D
    Figure imgb0068
    N-(2-{[(4-fluorobenzyl)amino]-carbonyl}-3-hydroxy-8,8-dimethyl-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide 474 C
    Figure imgb0069
    (+)-N-(2-{[(4-fluoro-3-methylbenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-Pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide 460 D
    Figure imgb0070
    N-(4-fluorobenzyl)-3-hydroxy-10-morpholin-4-yl-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxamide 417 B
    Figure imgb0071
    10-[[(dimethylamino)sulfonyl](methyl)-amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido-[1,2-a]azepine-2-carboxamide 468 C
    Figure imgb0072
    N-(4-fluorobenzyl)-3-hydroxy-10-{methyl[(methylsulfonyl)acetyl]amino}-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxamide 481 C
    Figure imgb0073
    8-[[(dimethylamino)sulfonyl](methyl)-amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-4,6,7,8-tetrahydropyrrolo[1,2-a]pyrimidine-2-carboxamido 440 C
    (Ex. 10)

Claims (10)

  1. A compound, or a pharmaceutically acceptable salt thereof, which is a compound of Formula II:
    Figure imgb0074
    R3 is hydrogen;
    R1 is NR2R5 ;
    R2 is CH3;
    R5 is
    1) C(O)CH2SO2CH3.
    2) C(O)C(O)N(CH3)2,
    3) SO2N(CH3)2, or
    4) SO2R20, wherein R20 is:
    Figure imgb0075
    or alternatively R2 and R5 together with the nitrogen atom to which they are attached form
    Figure imgb0076
    R4 is:
    1) p-fluorobenzyl,
    2) 4-fluoro-3-methylbenzyl,
    3) 3-chlorobenzyl, or
    4) 3-chloro-4-methylbenzyl;
    R12 and R14 are both H, except that when R5 is C(O)C(O)N(CH3)2 and R4 is p-fluorobenzyl and n is 1, then R12 and R14 are either both H or both CH3; and
    n is an integer equal to 1 or 2.
  2. A compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein
    R5 is C(O)C(O)N(CH3)2, or SO2R20, wherein R20 is
    Figure imgb0077
    R4 is p-fluorobenzyl or 4-fluoro-3-methylbenzyl;
    R12 and R14 are both H, except that when R5 is C(O)C(O)N(CH3)2 and R4 is p-fluorobenzyl and n is 1, then R12 and R14 are either both H or both CH3; and n is an integer equal to 1 or 2.
  3. A compound according to claim 1, or a pharmaceutically acceptable salt thereof, selected from the group consisting of:
    N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(methylsulfonyl)acetyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide:
    N-(4-fluorobenzyl)-3-hydroxy-9-morpholin-4-yl-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    N1-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N1,N2,N2-trimethylethanediamide;
    9-[[(dimethylamino)sulfonyl](methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]Pyrimidine-2-carboxamide:
    N-(4-fluorobenzyl)-3-hydroxy-9-{[(methylsulfonyl)acetyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide; (+)-9-[[(dimethylamino)sulfonyl](methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide:
    (-)-9-[[(dimethylamino)sulfonyl](methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    (-)N1-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N1,N2,N2-trimethylethanediamide;
    (+)N 1-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N1,N2,N2-trimethylethanediamide;
    (+)-N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(methylsulfonyl)acetyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide:
    (-)-N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(methylsulfonyl)acetyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    N-(2-{[(4-fluoro-3-methylbenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide;
    N-(2- [(3-chloro-4-methylbenzyl)amino]carbonyl-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide;
    N-(2-{[(3-chlorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide;
    N-(4-fluorobenzyl)-3-hydroxy-9-(6-methyl-1,1-dioxido-1,2,6-thiadiazinan-2-yl)-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    (-)-N-(2-{[(4-fluoro-3-methylbenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide;
    (-)-N-(2-{[(3-chloro-4-methylbenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide;
    (+)-N-(4-fluorobenzyl)-3-hydroxy-9-(6-methyl-1,1-dioxido-1,2,6-thiadiazinan-2-yl)-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    N-(4-fluorobenzyl)-3-hydroxy-9-[methyl(pyrrolidin-1-ylsulfonyl)amino]-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    9-[(azetidin-1-ylsulfonyl)(methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    (-)-N-(4-fluorobenzyl)-3-hydroxy-9-[methyl(morpholin-4-ylsulfonyl)amino]-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    (+)-N-(4-fluorobenzyl)-3-hydroxy-9-[methyl(morpholin-4-ylsulfonyl)amino]-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    (+)-9-[(azetidin-1-ylsulfonyl)(methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    (+)-N-(4-fluoro-3-methylbenzyl)-3-hydroxy-9-[methyl(morpholin-4-ylsulfonyl)amino]-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    (-)-N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(4-methylpiperazin-1-yl)sulfonyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin+2-carboxamide;
    (+)-N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(4-methylpiperazin-1-yl)sulfonyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(4-methylpiperazin-1-yl)sulfonyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    N-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide;
    (-)N-(2- {[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide;
    (+)N-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide;
    N-(2-{[(4-fluoro-3-methylbenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide;
    N-(2-{[(3-chloro-4-methylbenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide;
    (+)-N-(2- {[(4-fluoro-3-methylbenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide;
    (-)-N-(2-{[(4-fluoro-3-methylbenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide;
    N-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-8,8-dimethyl-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide;
    (+)-N-(2-{[(4-fluoro-3-methylbenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide;
    N-(4-fluorobenzyl)-3-hydroxy-10-morpholin-4-yl-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxamide;
    10-[[(dimethylamino)sulfonyl](methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxamide;
    N-(4-fluorobenzyl)-3-hydroxy-10-[methyl(methylsulfonyl)amino]-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxamide;
    N-(4-fluorobenzyl)-3-hydroxy-10-(6-methyl-1,1-dioxido-1,2,6-thiadiazinan-2-yl)-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepine-2-carboxamide; and 3-[[(dimethylamino)sulfonyl](methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-4,6,7,8-tetrahydropyrrolo[1,2-a]pyrimidine-2-carboxamide.
  4. A compound according to claim 3, or a pharmaceutically acceptable salt thereof, selected from the goup consisting of:
    (-)-N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(methylsulfonyl)acetyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    N-(2-{[(4-fluoro-3-methylbenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide;
    N-(2-{[(3-chloro-4-methylbenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide;
    N-(2-{[(3-chlorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide;
    N-(4-fluorobenzyl)-3-hydroxy-9-(6-methyl-1,1-dioxido-1,2,6-thiadiazinan-2-yl)-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    (-)-N-(2-{[(4-fluoro-3-methylbenzyl)amino]carbonyl}-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide;
    (+)-N-(4-fluorobenzyl)-3-hydroxy-9-(6-methyl-1,1-dioxido-1,2,6-thiadiazinan-2-yl)-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimdine-2-carboxamide;
    N-(4-fluorobenzyl)-3-hydroxy-9-[methyl(pyrrolidin-1-ylsulfonyl)amino]-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide:
    9-[(azetidin-1-ylsulfonyl)(methyl)amino]-N-(4-fluorobenzyl)3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    (-)-N-(4-fluorobenzyl)-3-hydroxy-9-[methyl(morpholin-4-ylsulfonyl)amino]-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    (+)-N-(4-fluorobenzyl)-3-hydroxy-9-[methyl(morpholin-4-ylsulfonyl)amino]-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    (+)-9-[(azetidin-1-ylsulfonyl)(methyl)amino]-N-(4-fluorobenzyl)-3-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    (-)-N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(4-methylpiperazin-1-yl)sulfonyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    (+)-N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(4-methylpiperazin-1-yl)sulfonyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    N-(4-fluorobenzyl)-3-hydroxy-9-{methyl[(4-methylpiperazin-1-yl)sulfonyl]amino}-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidine-2-carboxamide;
    N-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide;
    (-)N-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide;
    (+)N-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide;
    N-(2-{[(4-fluoro-3-methylbenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide;
    (+)-N-(2-{[(4-fluoro-3-methylbenzyl)amino]carbonyl -3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide:
    (-)-N-(2-{[(4-fluoro-3-methylbenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide; and N-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-8,8-dimethyl-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[1,2-a]pyrimidin-9-yl)-N,N',N'-trimethylethanediamide.
  5. A compound according to claim 4, or a pharmaceutically acceptable salt thereof, selected from the group consisting of:
    N-(2- {[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-1 0-yl)-N,N',N'-trimethylethanediamide;
    (-)N-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide; and
    (+)N-(2-{[(4-fluorobenzyl)amino]carbonyl}-3 -hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide.
  6. A compound according to claim 5, which is (-)N-(2-{[(4-fluorobenzyl)amino]carbonyl}-3-hydroxy-4-oxo-4,6,7,8,9,10-hexahydropyrimido[ 1,2-a]azepin-10-yl)-N,N',N'-trimethylethanediamide; or a pharmaceutically acceptable salt thereof.
  7. A pharmaceutical composition comprising a therapeutically effective amount of a compound according to any one of claims 1 to 6, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  8. A combination useful for treating or preventing infection by HIV, or for preventing, treating or delaying the onset of AIDS, which is a therapeutically effective amount of a compound according to any one of claims 1 to 6, or a pharmaceutically acceptable salt thereof, and a therapeutically effective amount of an an antiviral selected from the group consisting of HIV protease inhibitors, non-nucleoside HIV reverse transcriptase inhibitors and nucleoside HIV reverse transcriptase inhibitors.
  9. A compound according to any one of claims 1 to 6, or a pharmaceutically acceptable salt thereof, for use in therapy.
  10. Use of a compound according to any one of claims 1 to 6, or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for preventing or treating infection by HIV or for preventing, treating or delaying the onset of AIDS in a subject in need thereof.
EP03768014A 2002-12-27 2003-12-18 Tetrahydro-4h-pyrido[1,2-a]pyrimidines and related compounds useful as hiv integrase inhibitors Expired - Lifetime EP1578748B1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US43683002P 2002-12-27 2002-12-27
US436830P 2002-12-27
US52877603P 2003-12-12 2003-12-12
US528776P 2003-12-12
PCT/GB2003/005536 WO2004058756A1 (en) 2002-12-27 2003-12-18 Tetrahydro-4h-pyrido[1,2-a]pyrimidines and related compounds useful as hiv integrase inhibitors

Publications (2)

Publication Number Publication Date
EP1578748A1 EP1578748A1 (en) 2005-09-28
EP1578748B1 true EP1578748B1 (en) 2010-09-15

Family

ID=32685467

Family Applications (1)

Application Number Title Priority Date Filing Date
EP03768014A Expired - Lifetime EP1578748B1 (en) 2002-12-27 2003-12-18 Tetrahydro-4h-pyrido[1,2-a]pyrimidines and related compounds useful as hiv integrase inhibitors

Country Status (20)

Country Link
US (2) US7414045B2 (en)
EP (1) EP1578748B1 (en)
JP (1) JP4733986B2 (en)
KR (1) KR20050087865A (en)
CN (1) CN100343253C (en)
AT (1) ATE481401T1 (en)
AU (2) AU2003292436B2 (en)
BR (1) BR0317749A (en)
CA (1) CA2509554C (en)
DE (1) DE60334248D1 (en)
EC (1) ECSP055890A (en)
HR (1) HRP20050593A2 (en)
IS (1) IS7892A (en)
MA (1) MA27583A1 (en)
MX (1) MXPA05007010A (en)
NO (1) NO20053624L (en)
NZ (1) NZ540729A (en)
PL (1) PL377354A1 (en)
RU (1) RU2329265C2 (en)
WO (2) WO2004058756A1 (en)

Families Citing this family (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004062613A2 (en) * 2003-01-13 2004-07-29 Bristol-Myers Squibb Company Hiv integrase inhibitors
WO2005041664A1 (en) 2003-10-20 2005-05-12 Merck & Co., Inc. Hydroxy pyridopyrrolopyrazine dione compounds useful as hiv integrase inhibitors
AR046938A1 (en) * 2003-12-12 2006-01-04 Merck & Co Inc PROCEDURE TO PREPARE HEXAHYDROPIRIMID [1,2-A] AZEPIN-2-CARBOXYLATES AND SIMILAR COMPUTERS
TW200526635A (en) * 2003-12-22 2005-08-16 Shionogi & Co Hydroxypyrimidinone derivative having HIV integrase inhibitory activity
US7273859B2 (en) * 2004-05-12 2007-09-25 Bristol-Myers Squibb Company HIV integrase inhibitors: cyclic pyrimidinone compounds
US7115601B2 (en) 2004-05-18 2006-10-03 Bristol-Myers Squibb Company HIV integrase inhibitors
US7173022B2 (en) * 2004-05-28 2007-02-06 Bristol-Myers Squibb Company Bicyclic heterocycles as HIV integrase inhibitors
US7192948B2 (en) * 2004-05-28 2007-03-20 Bristol-Myers Squibb Company Bicyclic heterocycles as HIV integrase inhibitors
US7157447B2 (en) 2004-05-28 2007-01-02 Bristol-Myers Squibb Company Bicyclic heterocycles as HIV integrase inhibitors
US7176196B2 (en) 2004-05-28 2007-02-13 Bristol-Myers Squibb Company Bicyclic heterocycles as HIV integrase inhibitors
WO2006060225A2 (en) * 2004-11-23 2006-06-08 Merck & Co., Inc. Process for asymmetric synthesis of hexahydropyrimido[1,2-a] azepine-2-carboxamides and related compounds
CA2588465C (en) * 2004-12-03 2013-10-01 Merck & Co., Inc. Pharmaceutical composition containing an anti-nucleating agent
CN101068550B (en) 2004-12-03 2011-03-30 默沙东公司 Pharmaceutical formulation of carboxamide HIV integrase inhibitors containing a release rate controlling composition
KR20070085702A (en) * 2004-12-03 2007-08-27 머크 앤드 캄파니 인코포레이티드 Use of atazanavir for improving the pharmacokinetics of drugs metabolized by ugt1a1
UA87884C2 (en) * 2004-12-03 2009-08-25 Мерк Энд Ко., Инк. Potassium salt of an hiv integrase inhibitor
US7858788B2 (en) * 2005-02-21 2010-12-28 Shionogi & Co., Ltd. Bicyclic carbamoylpyridone derivative having HIV integrase inhibitory activity
CN101146811B (en) * 2005-03-31 2012-01-11 P.安杰莱蒂分子生物学研究所 Hiv integrase inhibitors
HUE044978T2 (en) * 2005-04-28 2019-11-28 Viiv Healthcare Co Polycyclic carbamoylpyridone derivative having hiv integrase inhibitory activity
AU2006266167A1 (en) 2005-06-30 2007-01-11 Wyeth Amino-5-(6-membered)heteroarylimidazolone compounds and the use thereof for beta-secretase modulation
US7494984B2 (en) * 2005-08-31 2009-02-24 Bristol-Myers Squibb Company Substituted imidazo[1,2-a]pyrimidines as HIV viral DNA integrase inhibitors
JP5094725B2 (en) * 2005-10-04 2012-12-12 イステイチユート・デイ・リチエルケ・デイ・ビオロジア・モレコラーレ・ピ・アンジエレツテイ・エツセ・エルレ・エルレ HIV integrase inhibitor
EA200801144A1 (en) 2005-10-27 2008-10-30 Сионоги Энд Ко., Лтд. POLYCYCLIC CARBAMOYLPYRIDON DERIVATIVE, HAS INHIBITOR ACTIVITY WITH RESPECT TO HIV INTEGRASES
EP1942736A2 (en) * 2005-10-27 2008-07-16 Merck & Co., Inc. Hiv integrase inhibitors
US7897592B2 (en) * 2005-11-15 2011-03-01 Bristol-Myers Squibb Company HIV integrase inhibitors
WO2007058646A1 (en) * 2005-11-15 2007-05-24 Bristol-Myers Squibb Company Hiv integrase inhibitors: cyclic pyrimidinone compounds
US7902182B2 (en) * 2005-11-16 2011-03-08 Bristol-Myers Squibb Company HIV integrase inhibitors
US8039458B2 (en) * 2005-11-17 2011-10-18 Bristol-Myers Squibb Company HIV integrase inhibitors
WO2007064316A1 (en) * 2005-11-30 2007-06-07 Bristol-Myers Squibb Company Bicyclic heterocycles as hiv integrase inhibitors
US20070129379A1 (en) * 2005-12-01 2007-06-07 Bristol-Myers Squibb Company Hiv integrase inhibitors
US20090136570A1 (en) * 2006-01-20 2009-05-28 Bhagwant Rege Taste-Masked Tablets and Granules
US7897593B2 (en) 2006-05-30 2011-03-01 Bristol-Myers Squibb Company HIV integrase inhibitors
US7893055B2 (en) * 2006-06-28 2011-02-22 Bristol-Myers Squibb Company HIV integrase inhibitors
US7763630B2 (en) * 2007-06-06 2010-07-27 Bristol-Myers Squibb Company HIV integrase inhibitors
US8129398B2 (en) * 2008-03-19 2012-03-06 Bristol-Myers Squibb Company HIV integrase inhibitors
EP2349276B1 (en) * 2008-10-06 2019-11-27 Merck Sharp & Dohme Corp. Hiv integrase inhibitors
US8624023B2 (en) * 2008-12-11 2014-01-07 Shionogi & Co., Ltd. Synthesis of carbamoylpyridone HIV integrase inhibitors and intermediates
US8143244B2 (en) * 2009-02-26 2012-03-27 Bristol-Myers Squibb Company Cyclopropyl fused indolobenzazepine HCV NS5B inhibitors
RU2561130C2 (en) * 2009-07-02 2015-08-20 Санофи NOVEL DERIVATIVES OF 1,2,3,4-TETRAHYDROPYRIMIDO{1,2-a}PYRIMIDIN-6-ONE, OBTAINING THEREOF AND PHARMACEUTICAL APPLICATION
CN102574854B (en) 2009-10-13 2014-04-16 伊兰科动物健康爱尔兰有限公司 Macrocyclic integrase inhibitors
US8383639B2 (en) 2009-10-15 2013-02-26 Bristol-Myers Squibb Company HIV integrase inhibitors
MY162494A (en) 2009-10-26 2017-06-15 Merck Sharp & Dohme Solid pharmaceutical compositions containing an integrase inhibitor
JP5739517B2 (en) 2010-04-02 2015-06-24 ヤンセン・アールアンドデイ・アイルランド Macrocyclic integrase inhibitor
UA112517C2 (en) 2010-07-06 2016-09-26 Новартіс Аг TETRAHYDROPYRIDOPYRIMIDINE DERIVATIVES
DK2790705T3 (en) 2011-12-15 2018-03-12 Novartis Ag Use of inhibitors of the activity or function of PI3K
WO2014099586A1 (en) 2012-12-17 2014-06-26 Merck Sharp & Dohme Corp. 4-pyridinonetriazine derivatives as hiv integrase inhibitors
EP2986291B1 (en) 2013-04-16 2020-05-27 Merck Sharp & Dohme Corp. 4-pyridone derivative compounds and uses thereof as hiv integrase inhibitors
ME02977B (en) 2013-05-17 2018-10-20 Merck Sharp & Dohme Fused tricyclic heterocyclic compounds as hiv integrase inhibitors
WO2014200880A1 (en) 2013-06-13 2014-12-18 Merck Sharp & Dohme Corp. Fused tricyclic heterocyclic compounds as hiv integrase inhibitors
KR102102516B1 (en) 2013-09-27 2020-04-20 머크 샤프 앤드 돔 코포레이션 Substituted quinolizine derivatives useful as hiv integrase inhibitors
WO2016187788A1 (en) 2015-05-25 2016-12-01 Merck Sharp & Dohme Corp. Fused tricyclic heterocyclic compounds useful for treating hiv infection
EP3377066B1 (en) 2015-11-17 2021-04-07 Merck Sharp & Dohme Corp. Amido-substituted pyridotriazine derivatives useful as hiv integrase inhibitors
WO2017106071A1 (en) 2015-12-15 2017-06-22 Merck Sharp & Dohme Corp. Spirocyclic quinolizine derivatives useful as hiv integrase inhibitors
WO2017113288A1 (en) 2015-12-31 2017-07-06 Merck Sharp & Dohme Corp. Fused tricyclic heterocyclic compounds as hiv integrase inhibitors
JOP20190130A1 (en) 2016-12-02 2019-06-02 Merck Sharp & Dohme Tetracyclic heterocycle compounds useful as hiv integrase inhibitors
CN110062627A (en) 2016-12-02 2019-07-26 默沙东公司 It can be used as the tricyclic heterocyclic compounds of hiv integrase inhibitor
US10786488B2 (en) 2017-01-26 2020-09-29 Merck Sharp & Dohme Corp. Substituted quinolizine derivatives useful as HIV integrase inhibitors
EA202092921A1 (en) 2018-05-31 2021-04-07 Сионоги Энд Ко., Лтд. POLYCYCLIC DERIVATIVE OF CARBAMOYLPIRIDONE
WO2019230857A1 (en) 2018-05-31 2019-12-05 塩野義製薬株式会社 Polycyclic pyridone derivative

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6262055B1 (en) 1998-06-03 2001-07-17 Merck & Co., Inc. HIV integrase inhibitors
US6380249B1 (en) 1998-06-03 2002-04-30 Merck & Co., Inc. HIV integrase inhibitors
US6306891B1 (en) 1998-06-03 2001-10-23 Merck & Co., Inc. HIV integrase inhibitors
CA2370500A1 (en) 1999-06-25 2001-01-04 Lekhanh O. Tran 1-(aromatic- or heteroaromatic-substituted)-3-(heteroaromatic substituted)-1,3-propanediones and uses thereof
IL152845A0 (en) 2000-06-07 2003-06-24 Vertex Pharma Fused heterocyclic compounds and pharmaceutical compositions containing the same
GB0017676D0 (en) 2000-07-19 2000-09-06 Angeletti P Ist Richerche Bio Inhibitors of viral polymerase
CA2321348A1 (en) * 2000-09-27 2002-03-27 Blaise Magloire N'zemba Aromatic derivatives with hiv integrase inhibitory properties
SK4322003A3 (en) 2000-10-12 2003-09-11 Merck & Co Inc Aza- and polyaza-naphthalenyl carboxamides useful as HIV integrase inhibitors
AU1532802A (en) 2000-10-12 2002-04-22 Merck & Co Inc Aza- and polyaza-naphthalenyl-carboxamides useful as hiv integrase inhibitors
ATE345129T1 (en) 2000-10-12 2006-12-15 Merck & Co Inc AZA- AND POLYAZA-NAPHTHALENYL CARBONIC ACID AMIDES AS HIV INTEGRASE INHIBITORS
US20050010048A1 (en) 2000-10-12 2005-01-13 Linghang Zhuang Aza-and polyaza-naphthalenly ketones useful as hiv integrase inhibitors
MXPA04000646A (en) 2001-08-10 2004-03-18 Shionogi & Co Antiviral agent.
CA2463975A1 (en) 2001-10-26 2003-05-01 Maria Emilia Di Francesco Dihydroxypyrimidine carboxamide inhibitors of hiv integrase
EA007060B1 (en) 2001-10-26 2006-06-30 Иституто Ди Ричерке Ди Биолоджиа Молеколаре П. Анджелетти Спа N-substituted hydroxypyrimidinone carboxamide inhibitors of hiv integrase
US7279487B2 (en) 2002-01-17 2007-10-09 Merck & Co., Inc. Hydroxynaphthyridinone carboxamides useful as HIV integrase inhibitors
WO2004004657A2 (en) 2002-07-09 2004-01-15 Bristol-Myers Squibb Company Hiv integrase inhibitors
WO2004062613A2 (en) 2003-01-13 2004-07-29 Bristol-Myers Squibb Company Hiv integrase inhibitors
US7037908B2 (en) 2003-04-24 2006-05-02 Bristol-Myers Squibb Company HIV integrase inhibitors
AR046938A1 (en) * 2003-12-12 2006-01-04 Merck & Co Inc PROCEDURE TO PREPARE HEXAHYDROPIRIMID [1,2-A] AZEPIN-2-CARBOXYLATES AND SIMILAR COMPUTERS

Also Published As

Publication number Publication date
US7968553B2 (en) 2011-06-28
AU2003292437A1 (en) 2004-07-22
ATE481401T1 (en) 2010-10-15
EP1578748A1 (en) 2005-09-28
WO2004058757A1 (en) 2004-07-15
WO2004058756A1 (en) 2004-07-15
CN1753892A (en) 2006-03-29
DE60334248D1 (en) 2010-10-28
AU2003292436B2 (en) 2009-07-30
PL377354A1 (en) 2006-01-23
CA2509554C (en) 2011-02-01
RU2005123807A (en) 2006-01-20
US7414045B2 (en) 2008-08-19
MXPA05007010A (en) 2005-08-18
HRP20050593A2 (en) 2006-03-31
ECSP055890A (en) 2005-09-20
US20080176869A1 (en) 2008-07-24
NO20053624L (en) 2005-09-26
NZ540729A (en) 2008-03-28
JP4733986B2 (en) 2011-07-27
AU2003292436A1 (en) 2004-07-22
MA27583A1 (en) 2005-10-03
CA2509554A1 (en) 2004-07-15
IS7892A (en) 2005-06-13
JP2006513200A (en) 2006-04-20
CN100343253C (en) 2007-10-17
BR0317749A (en) 2005-11-22
KR20050087865A (en) 2005-08-31
RU2329265C2 (en) 2008-07-20
NO20053624D0 (en) 2005-07-26
US20060046985A1 (en) 2006-03-02

Similar Documents

Publication Publication Date Title
EP1578748B1 (en) Tetrahydro-4h-pyrido[1,2-a]pyrimidines and related compounds useful as hiv integrase inhibitors
US7820680B2 (en) HIV integrase inhibitors
JP5116660B2 (en) HIV integrase inhibitor
US20070161639A1 (en) Hiv integrase inhibitors
NZ511015A (en) 1, 6-Dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one derivatives useful for treating impotence
WO2005087767A1 (en) Hiv integrase inhibitors
US20100216834A1 (en) Hiv integrase inhibitors
SK4322003A3 (en) Aza- and polyaza-naphthalenyl carboxamides useful as HIV integrase inhibitors
WO2002088080A2 (en) Dual inhibitors of pde 7 and pde 4
JP2008501021A (en) Bicyclic heterocycles as HIV integrase inhibitors
EP1937678B1 (en) Hiv integrase inhibitors
US20120022045A1 (en) Bridged compounds as hiv integrase inhibitors
WO2011025683A1 (en) Hiv integrase inhibitors
ZA200504853B (en) Tetrahydro-4h-Pyrido[1,2-A]Pyrimidines and related compound useful as HIV integrase inhibitors
AU2002256419A1 (en) Dual inhibitors of PDE 7 and PDE 4
UA74024C2 (en) A method for the treatment of states caused by ??38-kinases, and pyrrolotriazine compounds suitable for use as kinases inhibitors

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20050727

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK

RAX Requested extension states of the european patent have changed

Extension state: LT

Payment date: 20050727

Extension state: LV

Payment date: 20050727

17Q First examination report despatched

Effective date: 20070209

RTI1 Title (correction)

Free format text: TETRAHYDRO-4H-PYRIDO ??1, 2-A PYRIMIDINES AND RELATED COMPOUNDS USEFUL AS HIV INTEGRASE INHIBITORS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

RTI1 Title (correction)

Free format text: TETRAHYDRO-4H-PYRIDO(1,2-A)PYRIMIDINES AND RELATED COMPOUNDS USEFUL AS HIV INTEGRASE INHIBITORS

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 31/18 20060101ALI20071126BHEP

Ipc: C07D 487/04 20060101ALI20071126BHEP

Ipc: A61K 31/519 20060101ALI20071126BHEP

Ipc: C07D 471/04 20060101AFI20071126BHEP

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

APBK Appeal reference recorded

Free format text: ORIGINAL CODE: EPIDOSNREFNE

APBN Date of receipt of notice of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA2E

APBR Date of receipt of statement of grounds of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA3E

APBD Information on interlocutory revision deleted

Free format text: ORIGINAL CODE: EPIDOSDIRAPE

APBV Interlocutory revision of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNIRAPE

APBD Information on interlocutory revision deleted

Free format text: ORIGINAL CODE: EPIDOSDIRAPE

APBV Interlocutory revision of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNIRAPE

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: LT LV

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: SERVOPATENT GMBH

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

Ref country code: IE

Ref legal event code: FG4D

REF Corresponds to:

Ref document number: 60334248

Country of ref document: DE

Date of ref document: 20101028

Kind code of ref document: P

APAW Appeal reference deleted

Free format text: ORIGINAL CODE: EPIDOSDREFNO

APAY Date of receipt of notice of appeal deleted

Free format text: ORIGINAL CODE: EPIDOSDNOA2O

APBA Date of receipt of statement of grounds of appeal deleted

Free format text: ORIGINAL CODE: EPIDOSDNOA3O

APBE Information on interlocutory revision deleted

Free format text: ORIGINAL CODE: EPIDOSDIRAPO

APAW Appeal reference deleted

Free format text: ORIGINAL CODE: EPIDOSDREFNO

APAY Date of receipt of notice of appeal deleted

Free format text: ORIGINAL CODE: EPIDOSDNOA2O

APBM Appeal reference recorded

Free format text: ORIGINAL CODE: EPIDOSNREFNO

APBP Date of receipt of notice of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA2O

APBQ Date of receipt of statement of grounds of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNNOA3O

APBW Interlocutory revision of appeal recorded

Free format text: ORIGINAL CODE: EPIDOSNIRAPO

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20100915

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20100915

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IE

Payment date: 20101230

Year of fee payment: 8

LTIE Lt: invalidation of european patent or patent extension

Effective date: 20100915

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20100915

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20100915

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20101216

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20100915

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20100915

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20100915

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110117

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20100915

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20100915

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20100915

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20100915

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20101226

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20101231

26N No opposition filed

Effective date: 20110616

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20100915

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 60334248

Country of ref document: DE

Effective date: 20110616

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20100915

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20110316

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20101218

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20100915

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20111218

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20121026

Year of fee payment: 10

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20101215

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20131206

Year of fee payment: 11

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 60334248

Country of ref document: DE

Representative=s name: ABITZ & PARTNER, DE

REG Reference to a national code

Ref country code: GB

Ref legal event code: 732E

Free format text: REGISTERED BETWEEN 20140529 AND 20140604

REG Reference to a national code

Ref country code: DE

Ref legal event code: R081

Ref document number: 60334248

Country of ref document: DE

Owner name: MSD ITALIA S.R.L., IT

Free format text: FORMER OWNER: ISTITUTO DI RICERCHE DI BIOLOGIA MOLECOLARE P. ANGELETTI S.P.A., 00040 POMEZIA, IT

Effective date: 20140519

Ref country code: DE

Ref legal event code: R082

Ref document number: 60334248

Country of ref document: DE

Representative=s name: ABITZ & PARTNER, DE

Effective date: 20140519

Ref country code: DE

Ref legal event code: R082

Ref document number: 60334248

Country of ref document: DE

Representative=s name: ABITZ & PARTNER PATENTANWAELTE MBB, DE

Effective date: 20140519

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: FR

Ref legal event code: TP

Owner name: MSD ITALIA S.R.L., IT

Effective date: 20140827

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20131231

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20131231

REG Reference to a national code

Ref country code: NL

Ref legal event code: SD

Effective date: 20141031

REG Reference to a national code

Ref country code: NL

Ref legal event code: V1

Effective date: 20150701

REG Reference to a national code

Ref country code: NL

Ref legal event code: V1

Effective date: 20150701

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150701

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20151125

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20151124

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20151230

Year of fee payment: 13

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 60334248

Country of ref document: DE

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20161218

REG Reference to a national code

Ref country code: FR

Ref legal event code: ST

Effective date: 20170831

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170102

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20170701

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20161218